Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Brain Res Bull ; 155: 61-66, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31805305

RESUMO

Levodopa is a precursor to dopamine that has been shown to improve functional recovery following stroke partly achieved through mechanisms of brain plasticity. This study investigates if dopamine might affect plasticity by having a direct effect on synaptic plasticity through alterations in neurotransmitter release and re-uptake. Synaptogyrin is a synaptic vesicle protein that has been suggested to be involved in dopamine re-uptake in the synaptic terminal. Therefore, we investigated if levodopa has an effect on the expression of synaptogyrin 1. Thy1-YFP mice were subjected to photothrombosis as an experimental model of stroke. Starting two days after surgery they were treated with either levodopa or a vehicle solution (saline) on a daily basis until day seven following surgery. On day seven they were sacrificed and their brains stained for Dopamine 1 receptor (D1R), Dopamine 2 receptor (D2R) and Parvalbumin (PV). Neu-N stainings were used to estimate infarct size. A second group of mice were subjected to photothrombosis and also treated with either levodopa or a vehicle solution in the same manner as previously mentioned. On day seven they were then sacrificed, and samples of brain tissue were taken for protein determination. Western blots were carried out to investigate possible differences in synaptogyrin expression between the two groups. Immunofluorescent stains showed the presence of dopamine receptors on the YFP-positive neurons and on PV-expressing neurones. Our Western Blot analysis showed a significant decrease in the expression of synaptogyrin in levodopa-treated mice. Our stains showed co-localisation with Thy-1 neurones and PV-expressing neurones for both D1 and D2 receptors. This indicates that dopamine has the ability to bind to, and directly influence cortical neurons, as well as inhibitory interneurons. We discovered a considerable decrease in synaptogyrin expression through levodopa treatment, suggesting that this might be a mechanism for regulating brain plasticity.


Assuntos
Encéfalo/efeitos dos fármacos , Dopaminérgicos/administração & dosagem , Levodopa/administração & dosagem , Recuperação de Função Fisiológica , Acidente Vascular Cerebral/metabolismo , Sinaptogirinas/metabolismo , Animais , Encéfalo/metabolismo , Camundongos Transgênicos , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo
2.
Neuroimage Clin ; 17: 717-730, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29264113

RESUMO

Stroke causes direct structural damage to local brain networks and indirect functional damage to distant brain regions. Neuroplasticity after stroke involves molecular changes within perilesional tissue that can be influenced by regions functionally connected to the site of injury. Spontaneous functional recovery can be enhanced by rehabilitative strategies, which provides experience-driven cell signaling in the brain that enhances plasticity. Functional neuroimaging in humans and rodents has shown that spontaneous recovery of sensorimotor function after stroke is associated with changes in resting-state functional connectivity (RS-FC) within and across brain networks. At the molecular level, GABAergic inhibitory interneurons can modulate brain plasticity in peri-infarct and remote brain regions. Among this cell-type, a decrease in parvalbumin (PV)-immunoreactivity has been associated with improved behavioral outcome. Subjecting rodents to multisensory stimulation through exposure to an enriched environment (EE) enhances brain plasticity and recovery of function after stroke. Yet, how multisensory stimulation relates to RS-FC has not been determined. In this study, we investigated the effect of EE on recovery of RS-FC and behavior in mice after stroke, and if EE-related changes in RS-FC were associated with levels of PV-expressing neurons. Photothrombotic stroke was induced in the sensorimotor cortex. Beginning 2 days after stroke, mice were housed in either standard environment (STD) or EE for 12 days. Housing in EE significantly improved lost tactile-proprioceptive function compared to mice housed in STD environment. RS-FC in the mouse was measured by optical intrinsic signal imaging 14 days after stroke or sham surgery. Stroke induced a marked reduction in RS-FC within several perilesional and remote brain regions. EE partially restored interhemispheric homotopic RS-FC between spared motor regions, particularly posterior secondary motor. Compared to mice housed in STD cages, EE exposure lead to increased RS-FC between posterior secondary motor regions and contralesional posterior parietal and retrosplenial regions. The increased regional RS-FC observed in EE mice after stroke was significantly correlated with decreased PV-immunoreactivity in the contralesional posterior motor region. In conclusion, experimental stroke and subsequent housing in EE induces dynamic changes in RS-FC in the mouse brain. Multisensory stimulation associated with EE enhances RS-FC among distinct brain regions relevant for recovery of sensorimotor function and controlled movements that may involve PV/GABA interneurons. Our results indicate that targeting neural circuitry involving spared motor regions across hemispheres by neuromodulation and multimodal sensory stimulation could improve rehabilitation after stroke.


Assuntos
Isquemia Encefálica/fisiopatologia , Encéfalo/fisiopatologia , Recuperação de Função Fisiológica , Acidente Vascular Cerebral/fisiopatologia , Animais , Encéfalo/metabolismo , Isquemia Encefálica/complicações , Isquemia Encefálica/reabilitação , Mapeamento Encefálico , Meio Ambiente , Neurônios GABAérgicos/metabolismo , Camundongos Endogâmicos C57BL , Atividade Motora , Imagem Óptica , Parvalbuminas/metabolismo , Propriocepção , Acidente Vascular Cerebral/complicações , Reabilitação do Acidente Vascular Cerebral
3.
J Pharmacol Sci ; 127(1): 30-5, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25704015

RESUMO

The sigma-1 receptor (Sig-1R) is a single 25 kD polypeptide and a chaperone protein immersed in lipid rafts of the endoplasmic reticulum (ER) where it interacts with mitochondria at the mitochondria-associated ER membrane domain (MAM). Upon activation, the Sig-1R binds to the inositol trisphosphate receptor (IP3R), and modulates cellular calcium (Ca(2+)) homeostasis. Also, the activated Sig-1R modulates plasma membrane receptor and ion channel functions, and may regulate cellular excitability. Further, the Sig-1R promotes trafficking of lipids and proteins essential for neurotransmission, cell growth and motility. Activation of the Sig-1R provides neuroprotection and is neurorestorative in cellular and animal models of neurodegenerative diseases and brain ischaemia. Neuroprotection appears to be due to inhibition of cellular Ca(2+) toxicity and/or inflammation, and neurorestoration may include balancing abberant neurotransmission or stimulation of synaptogenesis, thus remodelling brain connectivity. Single nucleotide polymorphisms and mutations of the SIGMAR1 gene worsen outcome in Alzheimer's disease and myotrophic lateral sclerosis supporting a role of Sig-1R in neurodegenerative disease. The combined neuroprotective and neurorestorative actions of the Sig-1R, provide a broad therapeutic time window of Sig-1R agonists. The Sig-1R is therefore a strong therapeutic target for the development of new treatments for neurodegenerative diseases and stroke.


Assuntos
Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/fisiopatologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Receptores sigma/agonistas , Receptores sigma/fisiologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Humanos , Modelos Neurológicos , Mutação , Regeneração Nervosa/fisiologia , Receptores sigma/genética , Receptor Sigma-1
4.
Neurobiol Dis ; 65: 220-32, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24513087

RESUMO

Cerebral ischemia is characterized by an early disruption of GABAergic neurotransmission contributing to an imbalance of the excitatory/inhibitory equilibrium and neuronal death, but the molecular mechanisms involved are not fully understood. Here we report a downregulation of GABA(A) receptor (GABA(A)R) expression, affecting both mRNA and protein levels of GABA(A)R subunits, in hippocampal neurons subjected to oxygen-glucose deprivation (OGD), an in vitro model of ischemia. Similar alterations in the abundance of GABA(A)R subunits were observed in in vivo brain ischemia. OGD reduced the interaction of surface GABA(A)R with the scaffold protein gephyrin, followed by clathrin-dependent receptor internalization. Internalization of GABA(A)R was dependent on glutamate receptor activation and mediated by dephosphorylation of the ß3 subunit at serine 408/409. Expression of phospho-mimetic mutant GABA(A)R ß3 subunits prevented receptor internalization and protected hippocampal neurons from ischemic cell death. The results show a key role for ß3 GABA(A)R subunit dephosphorylation in the downregulation of GABAergic synaptic transmission in brain ischemia, contributing to neuronal death. GABA(A)R phosphorylation might be a therapeutic target to preserve synaptic inhibition in brain ischemia.


Assuntos
Glucose/deficiência , Hipóxia/patologia , Infarto da Artéria Cerebral Média/patologia , Neurônios/metabolismo , Neurônios/patologia , Receptores de GABA-B/metabolismo , Animais , Calpaína/farmacologia , Morte Celular/fisiologia , Células Cultivadas , Inibidores de Cisteína Proteinase/farmacologia , Modelos Animais de Doenças , Embrião de Mamíferos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Hipocampo/citologia , Humanos , Fosforilação/efeitos dos fármacos , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Ratos , Ratos Wistar , Receptores de GABA-B/genética , Fatores de Tempo
5.
Behav Brain Res ; 261: 282-8, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24406718

RESUMO

Acute treatment with 17ß-estradiol provides effective neuroprotection during the first days after acute brain injury, however, effects of chronic activation of estrogen receptor beta (ERß) on recovery of function after experimental stroke have not been investigated. The present study, therefore, was conducted to test if delayed treatment with the specific ERß ligand 4-(1-phenyl-cyclohexyl)-phenol (AC-131) improves recovery of lost neurological function after permanent focal stroke induced by photothrombosis in adult Sprague-Dawley rats. Treatment was initiated on day 2 after photothrombosis and AC-131 (1, 10, and 50 mg/kg) was administered by daily subcutaneous injections for 14 days. On day 2, 4, 6, 8, 11, 14, and 17 after photothrombosis, functional deficits were assessed by the paw placement test, a standardized grip strength test and an adhesive removal test. Daily treatment with AC-131 significantly improved test scores in all three behavioral tests. Importantly, improved function was not associated with a decrease in infarct volume on day 17 after stroke onset. Our results suggest that increased activity of the ERß is involved in mechanisms of stroke recovery.


Assuntos
Moduladores de Receptor Estrogênico/uso terapêutico , Receptor beta de Estrogênio/metabolismo , Recuperação de Função Fisiológica/fisiologia , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/etiologia , Animais , Infarto Encefálico/etiologia , Infarto Encefálico/patologia , Cicloexanos/farmacologia , Cicloexanos/uso terapêutico , Relação Dose-Resposta a Droga , Moduladores de Receptor Estrogênico/farmacologia , Trombose Intracraniana/complicações , Masculino , Força Muscular/efeitos dos fármacos , Fenóis/farmacologia , Fenóis/uso terapêutico , Desempenho Psicomotor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Estatísticas não Paramétricas , Fatores de Tempo
6.
PLoS One ; 7(9): e45118, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23028794

RESUMO

Activation of the sigma-1 receptor (Sig-1R) improves functional recovery in models of experimental stroke and is known to modulate microglia function. The present study was conducted to investigate if Sig-1R activation after experimental stroke affects mediators of the inflammatory response in the ischemic hemisphere. Male Wistar rats were subjected to transient occlusion of the middle cerebral artery (MCAO) and injected with the specific Sig-1R agonist 1-(3,4-dimethoxyphenethyl)-4-(3-phenylpropyl)piperazine dihydrochloride (SA4503) or saline for 5 days starting on day 2 after MCAO. Treatment did not affect the increased levels of the pro-inflammatory cytokines interleukin 1 beta (IL-1ß), tumor necrosis factor alpha (TNF-α), interferon gamma (IFN-γ), interleukin 4 (IL-4), interleukin 5 (IL-5), and interleukin 13 (IL-13) in the infarct core and peri-infarct area after MCAO. In addition, treatment with SA4503 did not affect elevated levels of nitrite, TNF-α and IL-1ß observed in primary cultures of microglia exposed to combined Hypoxia/Aglycemia, while the unspecific sigma receptor ligand 1,3-di-o-tolylguanidine (DTG) significantly decreased the production of nitrite and levels of TNF-α. Analysis of the ischemic hemisphere also revealed increased levels of ionized calcium binding adaptor molecule 1 (Iba1) levels in the infarct core of SA4503 treated animals. However, no difference in Iba1 immunoreactivity was detected in the infarct core. Also, levels of the proliferation marker proliferating cell nuclear antigen (PCNA) and OX-42 were not increased in the infarct core in rats treated with SA4503. Together, our results suggest that sigma-1 receptor activation affects Iba1 expression in microglia/macrophages of the ischemic hemisphere after experimental stroke but does not affect post-stroke inflammatory mediators.


Assuntos
Inflamação/tratamento farmacológico , Inflamação/etiologia , Piperazinas/farmacologia , Piperazinas/uso terapêutico , Receptores sigma/agonistas , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Antígeno CD11b/metabolismo , Citocinas/biossíntese , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/patologia , Inflamação/patologia , Inflamação/fisiopatologia , Mediadores da Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Nitritos/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ratos , Ratos Wistar , Receptores sigma/metabolismo , Recuperação de Função Fisiológica/efeitos dos fármacos , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia , Receptor Sigma-1
7.
Stroke ; 43(2): 507-13, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22096034

RESUMO

BACKGROUND AND PURPOSE: Delayed treatment of patients with stroke with levodopa/benserazide contributes to enhanced functional recovery, but the mechanisms involved are poorly understood. The present study was designed to investigate if levodopa/benserazide treatment improves recovery of lost neurological function and contributes to tissue reorganization in the rat brain after stroke. METHODS: Male Wistar rats were subjected to transient occlusion of the middle cerebral artery (120 minutes) and treated with levodopa (1, 5, and 20 mg/kg)/benserazide (15 mg/kg) or saline for 12 consecutive days starting on Day 2 after transient occlusion of the middle cerebral artery. Infarct volume was determined and sensorimotor function was assessed using the rotating pole test, a 28-point neuroscore, and a cylinder test on Days 2, 7, and 14 after transient occlusion of the middle cerebral artery. The spatiotemporal expression pattern of dopamine-1 and dopamine-2 receptors and the dopamine- and cAMP-regulated neuronal phosphoprotein in reactive astrocytes were analyzed in the ischemic hemisphere as well as in cultured astrocytes. RESULTS: Treatment with levodopa/benserazide significantly improved the recovery of sensorimotor function after transient occlusion of the middle cerebral artery without affecting the infarct volume. In addition, we found that different subpopulations of glial fibrillary acidic protein-positive astrocytes in the peri-infarct area express dopamine-1 receptors and dopamine-2 receptors as well as dopamine- and cAMP-regulated neuronal phosphoprotein. CONCLUSIONS: Our results strongly corroborate the concept of recovery enhancing actions of levodopa treatment after stroke. Also, astrocytes in the peri-infarct area may contribute to the dopamine enhanced recovery mechanisms.


Assuntos
Antiparkinsonianos/uso terapêutico , Benserazida/uso terapêutico , Levodopa/uso terapêutico , Recuperação de Função Fisiológica , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Astrócitos/metabolismo , Western Blotting , Hipóxia Celular , Células Cultivadas , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Combinação de Medicamentos , Imunofluorescência , Proteína Glial Fibrilar Ácida/metabolismo , Glucose/deficiência , Imuno-Histoquímica , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/patologia , Masculino , Ratos , Ratos Wistar , Receptores Dopaminérgicos/biossíntese , Acidente Vascular Cerebral/patologia
8.
J Neurosci ; 31(12): 4622-35, 2011 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-21430162

RESUMO

GABA is the major inhibitory neurotransmitter in the CNS and changes in GABAergic neurotransmission affect the overall activity of neuronal networks. The uptake of GABA into synaptic vesicles is mediated by the vesicular GABA transporter (VGAT), and changes in the expression of the transporter directly regulate neurotransmitter release. In this work we investigated the changes in VGAT protein levels during ischemia and in excitotoxic conditions, which may affect the demise process. We found that VGAT is cleaved by calpains following excitotoxic stimulation of hippocampal neurons with glutamate, giving rise to a stable truncated cleavage product (tVGAT). VGAT cleavage was also observed after transient middle cerebral artery occlusion in mice, a cerebral ischemia model, and following intrahippocampal injection of kainate, but no effect was observed in transgenic mice overexpressing calpastatin, a calpain inhibitor. Incubation of isolated cerebrocortical synaptic vesicles with recombinant calpain also induced the cleavage of VGAT and formation of stable tVGAT. Immunoblot experiments using antibodies targeting different regions of VGAT and N-terminal sequencing analysis showed that calpain cleaves the transporter in the N-terminal region, at amino acids 52 and 60. Immunocytochemistry of GABAergic striatal neurons expressing GFP fusion proteins with the full-length VGAT or tVGAT showed that cleavage of the transporter induces a loss of synaptic delivery, leading to a homogeneous distribution of the protein along neurites. Our results show that excitotoxicity downregulates full-length VGAT, with a concomitant generation of tVGAT, which is likely to affect GABAergic neurotransmission and may influence cell death during ischemia.


Assuntos
Neurotoxinas/farmacologia , Sinapses/metabolismo , Proteínas Vesiculares de Transporte de Glutamato/metabolismo , Animais , Western Blotting , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Calpaína/metabolismo , DNA/genética , Agonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Imuno-Histoquímica , Infarto da Artéria Cerebral Média/patologia , Ácido Caínico/farmacologia , Potenciação de Longa Duração/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Células PC12 , Monoéster Fosfórico Hidrolases/metabolismo , Plasmídeos/genética , Gravidez , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estado Epiléptico/metabolismo , Estado Epiléptico/patologia , Sinapses/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Transfecção , Ácido gama-Aminobutírico/fisiologia
9.
J Neuroinflammation ; 7: 94, 2010 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-21172031

RESUMO

BACKGROUND: Inflammation acting in synergy with brain ischemia aggravates perinatal ischemic brain damage. The sensitizing effect of pro-inflammatory exposure prior to hypoxia is dependent on signaling by TNF-α through TNF receptor (TNFR) 1. Adrenoceptor (AR) activation is known to modulate the immune response and synaptic transmission. The possible protective effect of α and ß AR activation against neuronal damage caused by tissue ischemia and inflammation, acting in concert, was evaluated in murine hippocampal organotypic slices treated with lipopolysaccharide (LPS) and subsequently subjected to oxygen-glucose deprivation (OGD). METHOD: Hippocampal slices from mice were obtained at P6, and were grown in vitro for 9 days on nitrocellulose membranes. Slices were treated with ß1(dobutamine)-, ß2(terbutaline)-, α1(phenylephrine)- and α2(clonidine)-AR agonists (5 and 50 µM, respectively) during LPS (1 µg/mL, 24 h) -exposure followed by exposure to OGD (15 min) in a hypoxic chamber. Cell death in the slice CA1 region was assessed by propidium iodide staining of dead cells. RESULTS: Exposure to LPS + OGD caused extensive cell death from 4 up to 48 h after reoxygenation. Co-incubation with ß1-agonist (50 µM) during LPS exposure before OGD conferred complete protection from cell death (P < 0.001) whereas the ß2-agonist (50 µM) was partially protective (p < 0.01). Phenylephrine was weakly protective while no protection was attained by clonidine. Exposure to both ß1- and ß2-agonist during LPS exposure decreased the levels of secreted TNF-α, IL-6 and monocyte chemoattractant protein-1 and prevented microglia activation in the slices. Dobutamine remained neuroprotective in slices exposed to pure OGD as well as in TNFR1-/- and TNFR2-/- slices exposed to LPS followed by OGD. CONCLUSIONS: Our data demonstrate that activation of both ß1- and ß2-receptors is neuroprotective and may offer mechanistic insights valuable for development of neuro-protective strategies in neonates.


Assuntos
Isquemia Encefálica/metabolismo , Encefalite/metabolismo , Glucose/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Lipopolissacarídeos/farmacologia , Oxigênio/metabolismo , Receptores Adrenérgicos beta/metabolismo , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Isquemia Encefálica/patologia , Morte Celular , Citocinas/metabolismo , Dobutamina/farmacologia , Encefalite/patologia , Hipocampo/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Fenilefrina/farmacologia , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Terbutalina/farmacologia , Técnicas de Cultura de Tecidos
10.
J Neurosci Res ; 88(15): 3414-23, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20890990

RESUMO

Deletion of the tumor suppressor gene p53 has been shown to improve the outcome in experimental models of focal cerebral ischemia and kainate-induced seizures. To evaluate the potential role of p53 in traumatic brain injury, genetically modified mice lacking a functional p53 gene (p53(-/-), n = 9) and their wild-type littermates (p53(+/+), n = 9) were anesthetized and subjected to controlled cortical impact (CCI) experimental brain trauma. After brain injury, neuromotor function was assessed by using composite neuroscore and rotarod tests. By 7 days posttrauma, p53(-/-) mice exhibited significantly improved neuromotor function, in the composite neuroscore (P = 0.002) as well as in two of three individual tests, when compared with brain-injured p53(+/+) animals. CCI resulted in the formation of a cortical cavity (mean volume = 6.1 mm(3)) 7 days postinjury in p53(+/+) as well as p53(-/-) mice. No difference in lesion volume was detected between the two genotypes (P = 0.95). Although significant cell loss was detected in the ipsilateral hippocampus and thalamus of brain-injured animals, no differences between p53(+/+) and p53(-/-) mice were detected. Although our results suggest that lack of the p53 gene results in augmented recovery of neuromotor function following experimental brain trauma, they do not support a role for p53 acting as a mediator of neuronal death in this context, underscoring the complexity of its role in the injured brain.


Assuntos
Lesões Encefálicas/genética , Lesões Encefálicas/patologia , Atividade Motora/fisiologia , Neurônios/patologia , Proteína Supressora de Tumor p53/genética , Animais , Camundongos , Camundongos Knockout , Recuperação de Função Fisiológica
11.
J Cereb Blood Flow Metab ; 30(10): 1756-66, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20234379

RESUMO

Various proteases in the brain contribute to ischemic brain injury. We investigated the involvement of the asparaginyl endopeptidase legumain after experimental stroke. On the basis of gene array studies and in situ hybridizations, we observed an increase of legumain expression in the peri-infarct area of rats after transient occlusion of the middle cerebral artery (MCAO) for 120 mins with a maximum expression at 24 and 48 h. Immunohistochemical analyses revealed the expression of legumain in Iba1(+) microglial cells and glial fibrillary acidic protein-positive astrocytes of the peri-infarct area in mice after MCAO. Post-stroke recovery was also studied in aged legumain-deficient mice (45 to 58 weeks old). Legumain-deficient mice did not show any differences in physiologic parameters compared with respective littermates before, during MCAO (45 mins), and the subsequent recovery period of 8 days. Moreover, legumain deficiency had no effect on mortality, infarct volume, and the neurologic deficit determined by the rotating pole test, a standardized grip strength test, and the pole test. However, a reduced number of invading CD74(+) cells in the ischemic hemisphere indicates an involvement in post-stroke inflammation. We conclude that legumain is not essential for the functional deficit after MCAO but may be involved in mechanisms of immune cell invasion.


Assuntos
Cisteína Endopeptidases/genética , Regulação da Expressão Gênica , Infarto da Artéria Cerebral Média/enzimologia , Acidente Vascular Cerebral/enzimologia , Animais , Antígenos de Diferenciação de Linfócitos B/imunologia , Astrócitos/imunologia , Encéfalo/citologia , Encéfalo/enzimologia , Encéfalo/patologia , Cisteína Endopeptidases/metabolismo , Antígenos de Histocompatibilidade Classe II/imunologia , Infarto da Artéria Cerebral Média/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Ratos Wistar , Acidente Vascular Cerebral/patologia
12.
J Cereb Blood Flow Metab ; 29(1): 73-86, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18728678

RESUMO

Inflammation and ischemia have a synergistic damaging effect in the immature brain. The role of tumor necrosis factor (TNF) receptors 1 and 2 in lipopolysaccharide (LPS)-induced sensitization and tolerance to oxygen-glucose deprivation (OGD) was evaluated in neonatal murine hippocampal organotypic slices. Hippocampal slices from balb/c, C57BL/6 TNFR1(-/-), TNFR2(-/-), and wild-type (WT) mice obtained at P6 were grown in vitro for 9 days. Preexposure to LPS immediately before OGD increased propidium iodide-determined cell death in regions CA1, CA3, and dentate gyrus from 4 up to 48 h after OGD (P<0.001). Extending the time interval between LPS exposure and OGD to 72 h resulted in tolerance, that is reduced neuronal cell death after OGD (P<0.05). Slices from TNFR1(-/-) mice showed neither LPS-induced sensitization nor LPS-induced tolerance to OGD, whereas both effects were present in slices from TNFR2(-/-) and WT mice. Cytokine secretion (TNFalpha and interleukin-6) during LPS exposure was decreased in TNFR1(-/-) slices and increased in TNFR2(-/-) as compared with WT slices. We conclude that LPS induces sensitization or tolerance to OGD depending on the time interval between exposure to LPS and OGD in murine hippocampal slice cultures. Both paradigms are dependent on signaling through TNFR1.


Assuntos
Glucose/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Lipopolissacarídeos/farmacologia , Oxigênio/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Animais , Animais Recém-Nascidos , Hipocampo/imunologia , Tolerância Imunológica/efeitos dos fármacos , Tolerância Imunológica/imunologia , Camundongos , Camundongos Knockout , Microglia/efeitos dos fármacos , Microglia/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/deficiência , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Fatores de Tempo , Técnicas de Cultura de Tecidos
13.
Curr Opin Neurobiol ; 16(3): 258-64, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16713245

RESUMO

Brain insults cause rapid cell death, and a disruption of functional circuits, in the affected regions. As the injured tissue recovers from events associated with cell death, regenerative processes are activated that over months lead to a certain degree of functional recovery. Factors produced by new neurons and glia, axonal sprouting of surviving neurons, and new synapse formation help to re-establish some of the lost functions. The timing and location of such events is crucial in the success of the regenerative process. Comprehensive gene expression profiling and proteomic analyses have enabled a deeper molecular and cellular mechanistic understanding of post-injury brain regeneration. These new mechanistic insights are aiding the design of novel therapeutic modalities that enhance regeneration.


Assuntos
Dano Encefálico Crônico/fisiopatologia , Lesões Encefálicas/fisiopatologia , Regeneração Nervosa/fisiologia , Plasticidade Neuronal/fisiologia , Recuperação de Função Fisiológica/fisiologia , Animais , Dano Encefálico Crônico/metabolismo , Dano Encefálico Crônico/terapia , Lesões Encefálicas/metabolismo , Lesões Encefálicas/terapia , Proliferação de Células , Cones de Crescimento/metabolismo , Humanos , Fatores de Crescimento Neural/metabolismo , Regeneração Nervosa/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Recuperação de Função Fisiológica/efeitos dos fármacos , Transplante de Células-Tronco/tendências
14.
Cell Transplant ; 14(5): 301-9, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16052911

RESUMO

Around 80-95% of the immature dopaminergic neurons die when embryonic ventral mesencephalic tissue is transplanted. Cell death occurs both during the preparation of donor tissue and after graft implantation, but the effect of combining successful neuroprotective treatments before and after transplantation has not been extensively investigated. We therefore treated embryonic rat mesencephalic tissue with a combination of the lipid peroxidation inhibitor tirilazad mesylate (3 microM) and the caspase inhibitor Ac.YVAD.cmk (500 microM) and transplanted the tissue into hemiparkinsonian rats kept hypothermic (32-33 degrees C) or normothermic (37 degrees C) during, and 90 min following, graft surgery. Suspension cell number did not differ between untreated or tirilazad/YVAD-treated preparations prior to transplantation. When graft survival was evaluated 6 weeks after implantation, both tirilazad/YVAD pretreatment and mild hypothermia increased the survival of transplanted dopaminergic neurons. Approximately 50-57% of the embryonic dopaminergic neurons survived the dissociation and grafting procedure in rats rendered hypothermic, but there was no significant additive effect on graft survival with a combined treatment. All groups of rats exhibited behavioral recovery in the amphetamine-induced rotation test. There was a significantly enhanced functional capacity of grafts placed in hypothermic as compared to normothermic rats. However, tirilazad/YVAD pretreated implants did not afford greater behavioral improvement than control-treated grafts. Our results suggest that neuroprotective treatments administered prior to and immediately after neural graft implantation may under certain conditions rescue, at least in part, the same subset of dopaminergic neurons. The study also emphasizes the importance of the immediate time after grafting for transplant survival, with relevance both for primary mesencephalic implants and stem cell grafts.


Assuntos
Clorometilcetonas de Aminoácidos/farmacologia , Hipotermia Induzida , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/transplante , Fármacos Neuroprotetores/farmacologia , Pregnatrienos/farmacologia , Animais , Inibidores de Caspase , Contagem de Células , Transplante de Células/métodos , Feminino , Sobrevivência de Enxerto/efeitos dos fármacos , Mesencéfalo/citologia , Mesencéfalo/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/transplante , Doença de Parkinson Secundária/terapia , Ratos , Ratos Sprague-Dawley , Transplante Heterólogo
15.
Brain Res ; 1049(1): 120-7, 2005 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-15935997

RESUMO

The aggravating effect of high glucose levels during cerebral ischemia has been extensively documented in clinical studies and in vivo models of global and focal ischemia. Detailed mechanistic studies of hyperglycemic ischemia have so far been hampered by the lack of in vitro models since glucose during anoxia in vitro is highly protective. We have previously reported glucose toxicity in murine hippocampal organotypic slice cultures exposed to anoxia in an acidotic medium containing high potassium and low calcium. In the present study, we compared the importance of calcium, nitric oxide and free radicals during in vitro ischemia (IVI) and hyperglycemic (40 mM) IVI. Extracellular calcium was a ubiquitous factor for cell death after IVI, but its removal from the medium had no effect on cell death after hyperglycemic IVI. When intracellular calcium was chelated by the 1,2-Bis(2-amino-5-fluorophenoxy)ethane-N,N,N',N'-tetraacetic acid tetrakis(acetoxymethyl) ester (BAPTA-AM) cell death appeared earlier but was mitigated in hyperglycemic IVI, while it was increased in glucose-free IVI. Addition of the nitric oxide synthase (NOS) inhibitor N(omega)-Nitro-L-arginine methyl ester hydrochloride (L-NAME) or the free radical scavengers N-tert-butyl-alpha-phenylnitrone (PBN), deferoxamine and N-acetyl-L-cysteine (NAC) did not affect cell damage in either paradigm. We conclude that the aggravating effect of hyperglycemia during in vitro ischemia is partially mediated by calcium ions released from intracellular stores.


Assuntos
Cálcio/metabolismo , Radicais Livres/metabolismo , Hipocampo/metabolismo , Hiperglicemia/metabolismo , Isquemia/metabolismo , Óxido Nítrico/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Quelantes/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Glucose/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Hiperglicemia/complicações , Hiperglicemia/patologia , Técnicas In Vitro , Líquido Intracelular/efeitos dos fármacos , Líquido Intracelular/metabolismo , Isquemia/etiologia , Isquemia/patologia , Camundongos , Camundongos Endogâmicos BALB C
16.
Eur J Neurosci ; 20(5): 1197-204, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15341591

RESUMO

Extracellular adenosine is dramatically increased during cerebral ischaemia and is considered to be neuroprotective due to its inhibitory effect on synaptic transmission mediated by the adenosine A1 receptor (A1R). We investigated the importance of the A1R in a mouse model of global ischaemia and in a murine hippocampal slice culture model of in vitro ischaemia, using mice with the A1R gene deleted. In brains from mice lacking the A1R, damage induced by global ischaemia was similar to that in wild-type animals. In contrast, treatment with a selective A1R antagonist [8-cyclo-pentyl theophylline (8-CPT)], administered before the ischaemic insult in naive wild-type mice, exacerbated the neuronal damage following global ischaemia. Although the inhibitory action of adenosine on excitatory neurotransmission in hippocampal slices was lost in A1R knockout mice, there was no difference in damage between slices from wild-type and knockout mice after in vitro ischaemia. The results suggest that some effects of the A1R are compensated for in knockout animals.


Assuntos
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Neurônios/metabolismo , Neurônios/patologia , Receptor A1 de Adenosina/deficiência , Receptor A1 de Adenosina/genética , Teofilina/análogos & derivados , Antagonistas do Receptor A1 de Adenosina , Animais , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Teofilina/farmacologia
17.
J Neurochem ; 85(6): 1431-42, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12787063

RESUMO

Blockade of mitochondrial permeability transition protects against hypoglycemic brain damage. To study the mechanisms downstream from mitochondria that may cause neuronal death, we investigated the effects of cyclosporin A on subcellular localization of apoptosis-inducing factor and cytochrome c, activation of the cysteine proteases calpain and caspase-3, as well as its effect on brain extracellular calcium concentrations. Redistribution of cytochrome c occurred at 30 min of iso-electricity, whereas translocation of apoptosis-inducing factor to nuclei occurred at 30 min of recovery following 30 min of iso-electricity. Active caspase-3 and calpain-induced fodrin breakdown products were barely detectable in the dentate gyrus and CA1 region of the hippocampus of rat brain exposed to 30 or 60 min of insulin-induced hypoglycemia. However, 30 min or 3 h after recovery of blood glucose levels, fodrin breakdown products and active caspase-3 markedly increased, concomitant with a twofold increase in caspase-3-like enzymatic activity. When rats were treated with neuroprotective doses of cyclosporin A, but not with FK 506, the redistribution of apoptosis-inducing factor and cytochrome c was reduced and fodrin breakdown products and active caspase-3 immuno-reactivity was diminished whereas the extracellular calcium concentration was unaffected. We conclude that hypoglycemia leads to mitochondrial permeability transition which, upon recovery of energy metabolism, mediates the activation of caspase-3 and calpains, promoting cell death.


Assuntos
Cálcio/metabolismo , Calpaína/metabolismo , Ciclosporina/farmacologia , Hipoglicemia/metabolismo , Neurônios/efeitos dos fármacos , Animais , Fator de Indução de Apoptose , Caspase 3 , Caspases/metabolismo , Contagem de Células , Grupo dos Citocromos c/metabolismo , Giro Denteado/citologia , Giro Denteado/metabolismo , Ativação Enzimática/efeitos dos fármacos , Flavoproteínas/metabolismo , Líquido Intracelular/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Microeletrodos , Neurônios/citologia , Neurônios/metabolismo , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Wistar , Tacrolimo/farmacologia
18.
J Cereb Blood Flow Metab ; 23(2): 219-31, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12571453

RESUMO

The cellular and molecular pathways initiated by traumatic brain injury (TBI) may compromise the function and structural integrity of mitochondria, thereby contributing to cerebral metabolic dysfunction and cell death. The extent to which TBI affects regional mitochondrial populations with respect to structure, function, and swelling was assessed 3 hours and 24 hours after lateral fluid-percussion brain injury in the rat. Significantly less mitochondrial protein was isolated from the injured compared with uninjured parietotemporal cortex, whereas comparable yields were obtained from the hippocampus. After injury, cortical and hippocampal tissue ATP concentrations declined significantly to 60% and 40% of control, respectively, in the absence of respiratory deficits in isolated mitochondria. Mitochondria with ultrastructural morphologic damage comprised a significantly greater percent of the population isolated from injured than uninjured brain. As determined by photon correlation spectroscopy, the mean mitochondrial radius decreased significantly in injured cortical populations (361 +/- 40 nm at 24 hours) and increased significantly in injured hippocampal populations (442 +/- 36 at 3 hours) compared with uninjured populations (Ctx: 418 +/- 44; Hipp: 393 +/- 24). Calcium-induced deenergized swelling rates of isolated mitochondrial populations were significantly slower in injured compared with uninjured samples, suggesting that injury alters the kinetics of mitochondrial permeability transition (MPT) pore activation. Cyclosporin A (CsA)-insensitive swelling was reduced in the cortex, and CsA-sensitive and CsA-insensitive swelling both were reduced in the hippocampus, demonstrating that regulated MPT pores remain in mitochondria isolated from injured brain. A proposed mitochondrial population model synthesizes these data and suggests that cortical mitochondria may be depleted after TBI, with a physically smaller, MPT-regulated population remaining. Hippocampal mitochondria may sustain damage associated with ballooned membranes and reduced MPT pore calcium sensitivity. The heterogeneous mitochondrial response to TBI may underlie posttraumatic metabolic dysfunction and contribute to the pathophysiology of TBI.


Assuntos
Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Mitocôndrias/patologia , Mitocôndrias/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Água Corporal/metabolismo , Cálcio/farmacologia , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Ciclosporina/farmacologia , Enzimas/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/ultraestrutura , Proteínas Mitocondriais/metabolismo , Consumo de Oxigênio , Permeabilidade/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA