Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 140
Filtrar
1.
Neuroscience ; 202: 334-41, 2012 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-22173016

RESUMO

Vascular endothelial growth factor (VEGF) is neuroprotective and induces neurogenesis and angiogenesis when given early after traumatic brain injury (TBI). However, the effects of VEGF administration in the subacute phase after TBI remain unknown. Mice were subjected to TBI and treated with vehicle or VEGF beginning 7 days later for an additional 7 days. The animals were injected with BrdU to label proliferating cells and examined with a motor-sensory scale at pre-determined time points. Mice were killed 90 days post injury and immunohistochemistry was used to study cell fates. Our results demonstrate that lesion volumes did not differ between the groups confirming the lack of neuroprotective effects in this paradigm. VEGF treatment led to significant increments in cell proliferation (1.9 fold increase vs. vehicle, P<0.0001) and angiogenesis in the lesioned cortex (1.7 fold increase vs. vehicle, P=0.0001) but most of the proliferating cells differentiated into glia and no mature newly-generated neurons were detected. In conclusion, VEGF induces gliogenesis and angiogenesis when given 7 days post TBI. However, treated mice had only insignificant motor improvements in this paradigm, suggesting that the bulk of the beneficial effects observed when VEGF is given early after TBI results from the neuroprotective effects.


Assuntos
Indutores da Angiogênese , Lesões Encefálicas/tratamento farmacológico , Neovascularização Fisiológica/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Antimetabólitos , Comportamento Animal/efeitos dos fármacos , Lesões Encefálicas/patologia , Lesões Encefálicas/psicologia , Bromodesoxiuridina , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Memória/efeitos dos fármacos , Camundongos , Reconhecimento Psicológico/efeitos dos fármacos , Resultado do Tratamento
2.
J Mol Neurosci ; 47(3): 546-53, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22127928

RESUMO

Preventing relapse to drug use is a major challenge for the treatment of drug addiction. Environmental cues are among the major determinants of relapse in abstinent cocaine users. The protein kinase M ζ (PKMζ) is involved in the generation and maintenance of long-term potentiation and is critical in memory storage. Here we show that inhibition of PKMζ in the nucleus accumbens (NAc) shell, a major component of the reward system that plays an important role in mediating drug craving and relapse, by a selective inhibitor ζ inhibitory peptide (ZIP), abolished cocaine-induced conditioned place preference (CPP). However, the injection of ZIP into the NAc core resulted in earlier onset of CPP extinction. Finally, we found that the levels of PKMζ and GluR2 in the NAc remained unchanged, while the GluR1 levels were elevated following CPP and fully reversed by ZIP injection. Together, our results suggest that inactivation of PKMζ in the NAc may result in the dissociation between the rewarding properties of the drug and the drug-related environment and may serve as a novel target for the treatment of drug relapse.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/tratamento farmacológico , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Peptídeos/farmacologia , Proteína Quinase C/antagonistas & inibidores , Recompensa , Animais , Condicionamento Psicológico/efeitos dos fármacos , Masculino , Núcleo Accumbens/enzimologia , Proteína Quinase C/metabolismo , Ratos , Ratos Endogâmicos
3.
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
4.
Neurobiol Dis ; 26(1): 86-93, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17270455

RESUMO

The mammalian target of rapamycin, commonly known as mTOR, is a serine/threonine kinase that regulates translation and cell division. mTOR integrates input from multiple upstream signals, including growth factors and nutrients to regulate protein synthesis. Inhibition of mTOR leads to cell cycle arrest, inhibition of cell proliferation, immunosuppression and induction of autophagy. Autophagy, a bulk degradation of sub-cellular constituents, is a process that keeps the balance between protein synthesis and protein degradation and is induced upon amino acids deprivation. Rapamycin, mTOR signaling inhibitor, mimics amino acid and, to some extent, growth factor deprivation. In the present study we examined the effect of rapamycin, on the outcome of mice after brain injury. Our results demonstrate that rapamycin injection 4 h following closed head injury significantly improved functional recovery as manifested by changes in the Neurological Severity Score, a neurobehavioral testing. To verify the activity of the injected rapamycin, we demonstrated that it inhibits p70S6K phosphorylation, reduces microglia/macrophages activation and increases the number of surviving neurons at the site of injury. We therefore suggest that rapamycin is neuroprotective following traumatic brain injury and as a drug used in the clinic for other indications, we propose that further studies on rapamycin should be conducted in order to consider it as a novel therapy for traumatic brain injury.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Fármacos Neuroprotetores , Sirolimo/uso terapêutico , Animais , Autofagia/efeitos dos fármacos , Western Blotting , Química Encefálica/efeitos dos fármacos , Lesões Encefálicas/patologia , Sobrevivência Celular/efeitos dos fármacos , Lateralidade Funcional/efeitos dos fármacos , Lateralidade Funcional/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Proteínas Quinases/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR
5.
Histol Histopathol ; 22(3): 235-50, 2007 03.
Artigo em Inglês | MEDLINE | ID: mdl-17163398

RESUMO

Traumatic brain injury causes progressive tissue atrophy and consequent neurological dysfunction, resulting from neuronal cell death in both animal models and patients. Fas (CD95) and Fas ligand (FasL/CD95L) are important mediators of apoptosis. However, little is known about the relationship between Fas and FasL and neuronal cell death in mice lacking the genes for inflammatory cytokines. In the present study, double tumor necrosis factor/lymphotoxin-alpha knockout (-/-) and interleukin-6-/- mice were subjected to closed head injury (CHI) and sacrificed at 24 hours or 7 days post-injury. Consecutive brain sections were evaluated for Fas and FasL expression, in situ DNA fragmentation (terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling; TUNEL), morphologic characteristics of apoptotic cell death and leukocyte infiltration. A peak incidence of TUNEL positive cells was found in the injured cortex at 24 hours which remained slightly elevated at 7 days and coincided with maximum Fas expression. FasL was only moderately increased at 24 hours and showed maximum expression at 7 days. A few TUNEL positive cells were also found in the ipsilateral hippocampus at 24 hours. Apoptotic, TUNEL positive cells mostly co-localized with neurons and Fas and FasL immunoreactivity. The amount of accumulated polymorphonuclear leukocytes and CD11b positive cells was maximal in the injured hemispheres at 24 hours. We show strong evidence that Fas and FasL might be involved in neuronal apoptosis after CHI. Furthermore, Fas and FasL upregulation seems to be independent of neuroinflammation since no differences were found between cytokine-/- and wild-type mice.


Assuntos
Lesões Encefálicas/metabolismo , Encéfalo/metabolismo , Proteína Ligante Fas/metabolismo , Ferimentos não Penetrantes/metabolismo , Receptor fas/metabolismo , Animais , Apoptose , Encéfalo/patologia , Lesões Encefálicas/patologia , Antígeno CD11b/metabolismo , Modelos Animais de Doenças , Técnica Indireta de Fluorescência para Anticorpo , Técnicas Imunoenzimáticas , Marcação In Situ das Extremidades Cortadas , Interleucina-6/deficiência , Interleucina-6/genética , Linfotoxina-alfa/deficiência , Linfotoxina-alfa/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Neurônios/patologia , Neutrófilos/patologia , Organismos Livres de Patógenos Específicos , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/genética , Regulação para Cima , Ferimentos não Penetrantes/patologia
6.
J Physiol Pharmacol ; 57 Suppl 11: 133-44, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17244945

RESUMO

Vascular endothelial cells are important not only for maintaining homeostasis, but also in pathogenesis of vascular disorders. Cerebral capillary and microvascular endothelial cells play an active role in maintaining cerebral blood flow, microvascular tone and blood brain barrier functions. Factors produced and released by endothelial cells, other brain cells and circulating blood cells participate in these regulatory functions. In particular, endothelin-1 (ET-1) and nitric oxide (NO) are known to contribute to the functional vascular changes under pathological conditions (e.g., hypertension, arteriosclerosis, and stroke). This report describes the involvement of endothelial cell mediators in the post-ischemic hypoperfusion induced by brain ischemia and in vitro endothelial responses (Ca(2+) mobilization and cytoskeletal rearrangements) to ET-1 and its interactions with NO or 2-AG. The capacity of NO and endocannabinoids to counteract ET-1-induced cerebral capillary and microvascular endothelial responses indicates that they may actively participate in EC function and implicates them in physiological and pathophysiological conditions.


Assuntos
Encéfalo/irrigação sanguínea , Endotelina-1/fisiologia , Endotélio Vascular/fisiopatologia , Actinas/metabolismo , Análise de Variância , Animais , Ácidos Araquidônicos/fisiologia , Barreira Hematoencefálica , Isquemia Encefálica/etiologia , Isquemia Encefálica/fisiopatologia , Cálcio/metabolismo , Células Cultivadas , Citoesqueleto/metabolismo , Endocanabinoides , Endotelina-1/metabolismo , Endotélio Vascular/metabolismo , Gerbillinae , Glicerídeos/fisiologia , Humanos , Imuno-Histoquímica , Microcirculação , Óxido Nítrico/fisiologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/fisiopatologia
7.
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
8.
Neuroimage ; 20(4): 1971-81, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14683703

RESUMO

Increases in peripheral type benzodiazepine receptors (PTBR) have been utilized for the detection of neuroinflammation and neurotoxicity in the brain. We have investigated the relationship between PTBR and NMDA receptor binding density in mice with closed head injury (CHI) using quantitative autoradiography. CHI was induced by a weight drop in nine mice, four of which received a single injection of the rat sarcoma (Ras) inhibitor famesyl thiosalicylate (FTS) 1 h after the insult. Sham controls received anesthesia but no contusion. The neurological status of the mice was evaluated at 1 h, and hence up to 7 days using a neurological severity score (NSS). Animals were killed 7 days after CHI and consecutive brain sections were incubated with [3H]PK11195, a PTBR antagonist, or [3H]MK801, an n-methyl-d-aspartate receptor (NMDAR) use-dependent antagonist. CHI produced large (two- to threefold), widespread increases in PK11195 binding in the traumatized hemisphere and a significant decrease (20%-40%) in NMDAR binding limited to regions at close proximity to the lesion. Histologically, these regions were characterized by glial proliferation and neuronal loss. Significant increases in PTBR binding, but no concomitant decrease in NMDAR, were identified in several regions remote from the lesion, including the contralateral ventrolateral striatum and the ipsilateral ventral thalamus. Drug treatment significantly improved the neurological deficits but had only a marginal effect on PTBR. These results support a complex role for glial activation and PTBR increases in the context of CHI.


Assuntos
Traumatismos Cranianos Fechados/fisiopatologia , Receptores de GABA-A/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Antioxidantes/farmacologia , Autorradiografia , Benzoatos/farmacologia , Encéfalo/patologia , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Farneseno Álcool/análogos & derivados , Farneseno Álcool/farmacologia , Ácido Glutâmico/fisiologia , Traumatismos Cranianos Fechados/metabolismo , Traumatismos Cranianos Fechados/patologia , Interpretação de Imagem Assistida por Computador , Isoquinolinas/farmacologia , Camundongos , Neuroglia/metabolismo , Receptores de GABA-A/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Compostos de Sulfidrila , Timerosal
9.
Neuroscience ; 121(2): 315-25, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14521991

RESUMO

Apolipoprotein E (apoE4) and head trauma are important genetic and environmental risk factors for Alzheimer's disease. Furthermore, apoE4 increases both the acute and chronic consequences of head trauma. The latter are associated with the deposition of amyloid-beta, which is particularly elevated in apoE4 subjects. The short-term effects of head injury are associated with transiently increased metabolism of amyloid precursor protein (APP) and its secreted fragment, APPs. In the present study, we examined the possibility that the acute, short-term pathological effects of apoE4 following head trauma and the corresponding neuroprotective effects of apoE3 are related to isoform-specific effects of apoE on APP metabolism. Accordingly, male transgenic mice expressing human apoE3 or apoE4 on a null mouse apoE background and apoE-deficient and control mice were subjected to closed head injury (CHI). The resulting effects on brain APP, and on its secreted products, APPs and secreted product of the alpha-cleavage of APP (APPsalpha) were then determined 24 h following injury. Immunoblotting revealed no significant differences between the basal APP, APPs and APPsalpha levels of the hippocampus or the cortex of the control and the apoE3 and ApoE4 transgenic mice. The apoE-deficient mice also had similar cortical basal levels of APP and its metabolites, whereas their corresponding basal hippocampal APP and APPs levels were lower than those of the other groups. CHI lowered the hipppocampal APPs and APPsalpha levels of the apoE4 transgenic mice, whereas those of the apoE3 transgenic mice and of the control and apoE-deficient mice were not affected by this insult. In contrast, CHI raised the cortical APP and APPs levels of the apoE3 transgenic mice but had no significant effect on those of the other mice groups. These animal model findings suggest that the acute, short-term pathological effects of apoE4 following CHI and the corresponding neuroprotective effects of apoE3 may be mediated by their opposing effects on the expression and cleavage of cortical and hippocampal APP. Similar isoform-specific interactions between apoE and APP may play a role in the acute, short-term effects of head trauma in humans.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Apolipoproteínas E/metabolismo , Traumatismos Cranianos Fechados/metabolismo , Animais , Apolipoproteína E3 , Apolipoproteína E4 , Apolipoproteínas E/deficiência , Química Encefálica , Modelos Animais de Doenças , Humanos , Immunoblotting , Masculino , Camundongos , Camundongos Transgênicos , Frações Subcelulares/metabolismo
10.
Acta Neurochir Suppl ; 86: 271-5, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14753451

RESUMO

Reactive oxygen species (ROS) were shown to play a role in altering blood-brain barrier (BBB) permeability and formation of brain edema induced by trauma and/or ischemia. 2-arachidonoyl glycerol (2-AG), a novel, potent vasodilatory and cytoprotective endocannabinoid has been implicated to act as an antioxidative agent. This study examines: 1) the possible 2-AG modulation of BBB injury and edema formation induced by closed head injury (CHI); and 2) comparable effects between 2-AG and 4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl (TPL), a known antioxidant nitroxide on endothelial Ca2+ and cytoskeletal responses to H2O2 (ROS). 2-AG treatment reduced the CHI-induced increase in BBB permeability and brain edema. The endothelial H2O2-stimulated Ca2+ mobilization and cytoskeleton (vimentin) rearrangement was modified by either 2-AG or TPL. These findings provide evidence of 2-AG antioxidant activity and are consistent with the involvement of ROS in the pathomechanism of CHI-induced BBB injury and brain edema.


Assuntos
Antioxidantes/farmacologia , Ácidos Araquidônicos/farmacologia , Barreira Hematoencefálica , Edema Encefálico/etiologia , Moduladores de Receptores de Canabinoides/farmacologia , Endocanabinoides , Glicerídeos/farmacologia , Traumatismos Cranianos Fechados/complicações , Traumatismos Cranianos Fechados/fisiopatologia , Sistema Vasomotor/efeitos dos fármacos , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Cálcio/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Óxidos N-Cíclicos/farmacologia , Citoesqueleto/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Sequestradores de Radicais Livres/farmacologia , Peróxido de Hidrogênio/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos , Oxidantes/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Marcadores de Spin
11.
Acta Neurochir Suppl ; 83: 49-54, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12442621

RESUMO

Traumatic brain injury is one of the most common causes for chronic disability in young people. Despite this there are currently no widely available modes of therapy that would limit the extent of brain damage secondary to trauma. Therefore, new insights into the pathological mechanisms involved in head trauma possibly leading to the identification of new therapeutic targets are urgently needed. In order to attain these goals adequate animal models for traumatic brain injury are needed. In the following paper the authors will review the various animal models for head trauma and emphasize their potential strengths and weaknesses.


Assuntos
Lesões Encefálicas/fisiopatologia , Modelos Animais de Doenças , Animais , Concussão Encefálica/fisiopatologia , Concussão Encefálica/terapia , Lesões Encefálicas/terapia , Córtex Cerebral/lesões , Córtex Cerebral/fisiopatologia , Traumatismos Cranianos Fechados/fisiopatologia , Traumatismos Cranianos Fechados/terapia , Humanos , Camundongos , Ratos , Fraturas Cranianas/fisiopatologia , Fraturas Cranianas/terapia
12.
Exp Neurol ; 176(2): 355-63, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12359177

RESUMO

Free radicals appear to participate in the final common pathway of neuronal death in ischemia and may therefore be an adequate target for therapy. Tempol is a nitroxide antioxidant with proven protective efficacy in several animal models, including myocardial ischemia, that has not been previously tested in models of permanent cerebral ischemia. Spontaneously hypertensive rats underwent permanent middle cerebral artery occlusion (PMCAO). Following dose-response and time-window-finding experiments rats were given vehicle or tempol (50 mg/kg) subcutaneously 1 h after PMCAO (n = 10/group). Five animals in each group were evaluated with a motor scale 24 h after the infarct and were then sacrificed and the injury volume was measured. The remaining animals were examined daily with the motor scale and also with a Morris water maze test on days 26-30 after PMCAO and sacrificed on day 30. Motor scores at all time points examined were significantly better in the tempol-treated animals (P < 0.05 for all). Significantly better performance in the water maze test for performance on days 26-30 was noted in the tempol group compared with the vehicle-treated group (P < 0.05). Injury volumes at days 1 and 30 were significantly reduced in the tempol group (9.83 +/- 1.05 vs 19.94 +/- 1.43% hemispheric volume, P = 0.0009, and 13.2 +/- 2.97 vs 24.4 +/- 2.38% hemispheric volume, P = 0.02, respectively). In conclusion, treatment with tempol led to significant motor and behavioral improvement and reduced injured tissue volumes both in the short and in the long term after stroke.


Assuntos
Antioxidantes/uso terapêutico , Isquemia Encefálica/tratamento farmacológico , Óxidos N-Cíclicos/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Tirosina/análogos & derivados , Animais , Comportamento Animal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Química Encefálica , Isquemia Encefálica/etiologia , Isquemia Encefálica/patologia , Modelos Animais de Doenças , Progressão da Doença , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/patologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos , Ratos , Ratos Endogâmicos SHR , Marcadores de Spin , Resultado do Tratamento , Tirosina/análise
13.
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
14.
Neurosci Lett ; 316(3): 169-72, 2001 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-11744229

RESUMO

Reactive oxygen species (ROS) are a major cause of secondary brain injury following head trauma. Low molecular weight antioxidants (LMWA) protect the tissue against oxidative damage caused by ROS. In the present study, we measured the extracellular levels of the LMWA ascorbic acid and uric acid in the rat brain before, during and after experimental closed head injury (CHI). A dialysis probe was inserted into the right ventral hippocampus through a chronically implanted guide. CHI was applied to the left hemisphere using a weight-drop device. CHI induced a rapid but transient increase in ascorbic acid levels. Uric acid levels increased to 250% of baseline shortly after CHI and remained elevated at 2 h after CHI. Previous results show that the overall reducing power of brain tissue decreases following CHI. Together with previous results, the current findings suggest that ascorbic acid and uric acid are mobilized from brain cells to the extracellular space.


Assuntos
Antioxidantes/metabolismo , Lesões Encefálicas/metabolismo , Espaço Extracelular/metabolismo , Traumatismos Cranianos Fechados/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Regulação para Cima/fisiologia , Animais , Ácido Ascórbico/metabolismo , Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Traumatismos Cranianos Fechados/patologia , Traumatismos Cranianos Fechados/fisiopatologia , Hipocampo/patologia , Hipocampo/fisiopatologia , Masculino , Microdiálise , Neurônios/patologia , Estresse Oxidativo/fisiologia , Ratos , Ratos Endogâmicos , Espécies Reativas de Oxigênio/metabolismo , Ácido Úrico/metabolismo
15.
Nature ; 413(6855): 527-31, 2001 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-11586361

RESUMO

Traumatic brain injury triggers the accumulation of harmful mediators that may lead to secondary damage. Protective mechanisms to attenuate damage are also set in motion. 2-Arachidonoyl glycerol (2-AG) is an endogenous cannabinoid, identified both in the periphery and in the brain, but its physiological roles have been only partially clarified. Here we show that, after injury to the mouse brain, 2-AG may have a neuroprotective role in which the cannabinoid system is involved. After closed head injury (CHI) in mice, the level of endogenous 2-AG was significantly elevated. We administered synthetic 2-AG to mice after CHI and found significant reduction of brain oedema, better clinical recovery, reduced infarct volume and reduced hippocampal cell death compared with controls. When 2-AG was administered together with additional inactive 2-acyl-glycerols that are normally present in the brain, functional recovery was significantly enhanced. The beneficial effect of 2-AG was dose-dependently attenuated by SR-141761A, an antagonist of the CB1 cannabinoid receptor.


Assuntos
Ácidos Araquidônicos , Lesões Encefálicas , Canabinoides , Glicerídeos/fisiologia , Fármacos Neuroprotetores/farmacologia , Animais , Temperatura Corporal , Edema Encefálico/etiologia , Edema Encefálico/prevenção & controle , Infarto Encefálico/prevenção & controle , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Canabinoides/metabolismo , Morte Celular , Modelos Animais de Doenças , Endocanabinoides , Cromatografia Gasosa-Espectrometria de Massas , Glicerídeos/administração & dosagem , Glicerídeos/farmacologia , Traumatismos Cranianos Fechados/metabolismo , Hipocampo/patologia , Masculino , Camundongos , Fármacos Neuroprotetores/administração & dosagem , Receptores de Canabinoides , Receptores de Droga/metabolismo
16.
Pediatr Dent ; 23(4): 337-42, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11572493

RESUMO

PURPOSE: Effective verbal communication is essential for successful dental treatment. The purpose of this study was to qualitatively and quantitatively examine communication techniques used by pediatric postdoctoral students during treatment. The operation of a communication model comprised of three linguistic approaches-permissive, empathic and personal-together with other strategies common to all three, was examined. METHODS: The study group consisted of 24 children (14 boys and 10 girls), 3 to 12 years of age. All four dentists were 2nd year residents in pediatric dentistry. Conversations were taped and analyzed linguistically and statistically. The frequency of use of each approach was tabulated and correlated to the children's reported anxiety, cooperation during treatment, success of treatment, and mood at the end of treatment. RESULTS: All dentists used the three approaches; the permissive approach, which supplied procedural information, was the most frequently used approach. The empathic approach was the least frequently used. Correlation tests showed that the empathic approach was most significantly related to the success of the treatment. Components of the permissive approach that contributed to the success of treatment were sensory information and supplying reasons. CONCLUSIONS: Although generalization is limited because of the small sample, improving verbal conversational skills, emphasizing certain strategies, and improving linguistic abilities will contribute to better communication between child and pediatric dentist and to better cooperation and success in treatment.


Assuntos
Comunicação , Relações Dentista-Paciente , Afeto , Assertividade , Terapia Aversiva , Criança , Comportamento Infantil , Pré-Escolar , Comportamento Cooperativo , Ansiedade ao Tratamento Odontológico/psicologia , Assistência Odontológica para Crianças , Empatia , Feminino , Humanos , Internato e Residência , Relações Interpessoais , Linguística , Masculino , Modelos Psicológicos , Comunicação não Verbal , Educação de Pacientes como Assunto , Odontopediatria/educação , Comunicação Persuasiva , Projetos Piloto , Reforço Psicológico , Estatística como Assunto , Resultado do Tratamento
17.
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
18.
Neuroreport ; 12(9): 2059-64, 2001 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-11435946

RESUMO

The expression of the chemokines macrophage inflammatory protein (MIP)-2 and MIP-1alpha and of their receptors CXCR2 and CCR5 was assessed in wild type (WT) and TNF/lymphotoxin-alpha knockout (TNF/LT-alpha-/-) mice subjected to closed head injury (CHI). At 4 h after trauma intracerebral MIP-2 and MIP-1alpha levels were increased in both groups with MIP-2 concentrations being significantly higher in WT than in TNF/LT-alpha-/- animals (p < 0.05). Thereafter, MIP-2 production declined rapidly, whereas MIP-1alpha remained elevated for 7 days. Expression of CXCR2 was confined to astrocytes and increased dramatically within 24 h in both mouse types. Contrarily, CCR5 expression remained constitutively low and was mainly localized to microglia. These results show that after CHI, chemokines and their receptors are regulated differentially and with independent kinetics.


Assuntos
Córtex Cerebral/metabolismo , Quimiocinas/metabolismo , Encefalite/metabolismo , Traumatismos Cranianos Fechados/metabolismo , Receptores de Quimiocinas/metabolismo , Animais , Astrócitos/metabolismo , Córtex Cerebral/fisiopatologia , Quimiocina CCL3 , Quimiocina CCL4 , Quimiocina CXCL2 , Encefalite/fisiopatologia , Regulação da Expressão Gênica/fisiologia , Traumatismos Cranianos Fechados/fisiopatologia , Linfotoxina-alfa/genética , Linfotoxina-alfa/metabolismo , Proteínas Inflamatórias de Macrófagos/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Monocinas/metabolismo , Receptores CCR5/metabolismo , Receptores de Interleucina-8B/metabolismo , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/genética , Regulação para Cima/genética
19.
J Mol Neurosci ; 16(1): 33-9, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11345518

RESUMO

We examined in the present investigation regional ATP, glucose, and lactate content in the cortical and subcortical structures, in a rat model of closed head injury (CHI). In serial tissue sections bioluminescence imaging of ATP, glucose, and lactate was performed at 4 h, 12 h and 24 h (n = 4/5 per time point with) after the induction of CHI or sham surgery. Bioluminescence images were analyzed by computer-assisted densitometry, at the lesion site, in remote cortical areas, and in the subcortical structures (thalamus and caudate nucleus). ATP content was significantly decreased at the lesion site after 4 h and in the remote cortex at 12 h post-injury. At 12 h, the ATP content reached baseline levels on the ipsilateral side and at 24 h also at remote lateral parietal sites. In the contralateral cortex, ATP increased transiently above the baseline at 12 h. No significant changes in ATP were found in the thalamus and caudate nucleus. Cortical glucose and lactate contents could not be discerned over time. Following CHI there is an acute and progressive, yet transient, ischemic cortical profile, which is not reflected in subcortical areas.


Assuntos
Lesões Encefálicas/metabolismo , Encéfalo/metabolismo , Encefalite/metabolismo , Metabolismo Energético/fisiologia , Neurônios/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Encéfalo/patologia , Encéfalo/fisiopatologia , Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Encefalite/fisiopatologia , Lateralidade Funcional/fisiologia , Glucose/metabolismo , Ácido Láctico/metabolismo , Medições Luminescentes , Masculino , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
20.
Brain Res ; 901(1-2): 195-201, 2001 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-11368967

RESUMO

In order to test the long-term cerebroprotective effects of dexanabinol, a synthetic non-competitive NMDA antagonist that also has anti-TNFalpha effects, spontaneously hypertensive rats underwent permanent middle cerebral artery occlusion (PMCAO). Rats were given vehicle or dexanabinol (4.5 mg/kg) 1, 3 or 6 h after PMCAO. The research consisted of 2 stages. In the short-term set of experiments animals (n=5/group), were tested with a motor disability scale 24 h post PMCAO, then sacrificed and the infarct volume was measured using 2,3,5-Triphenyltetrazolium chloride (TTC) staining. In the long-term set of experiments the rats (n=7/group) were examined daily with a motor disability scale up to 30 days after PMCAO and then sacrificed and infarct volumes were determined using TTC staining. Motor scores were significantly improved in the dexanabinol treated rats (P<0.05 for all groups) at all the time points examined. Infarct volumes were significantly reduced 24 h after PMCAO in the groups treated 1 or 3 h, but not 6 h after PMCAO compared with vehicle (Mean+/-S.D., 11.5+/-2.02, 12+/-3.2 and 14.4+/-2.4% vs. 20.8+/-1.3% hemispheric volume respectively). The lesions remained significantly smaller in the dexanabinol groups 30 days after PMCAO (Mean+/-S.D., 24.49+/-1.9% vs. 8.1+/-0.6, 11.1+/-2.3 and 13.8+/-2.5% hemispheric volume in animals treated with vehicle vs. dexanabinol 1, 3 or 6 h after PMCAO respectively; P<0.05 for all). In conclusion, the extended therapeutic window and the multi-mechanistic durable neuroprotective effects of dexanabinol make it a promising candidate for future stroke therapy.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Córtex Cerebral/efeitos dos fármacos , Dronabinol/análogos & derivados , Dronabinol/farmacologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Fenômenos Fisiológicos Cardiovasculares/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/fisiopatologia , Modelos Animais de Doenças , Esquema de Medicação , Encefalite/tratamento farmacológico , Encefalite/etiologia , Encefalite/prevenção & controle , Radicais Livres/antagonistas & inibidores , Radicais Livres/metabolismo , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Transtornos dos Movimentos/tratamento farmacológico , Transtornos dos Movimentos/etiologia , Transtornos dos Movimentos/fisiopatologia , Degeneração Neural/tratamento farmacológico , Degeneração Neural/etiologia , Degeneração Neural/prevenção & controle , Neurônios/metabolismo , Neurônios/patologia , Ratos , Ratos Endogâmicos SHR , Sais de Tetrazólio/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...