Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Crit Care Med ; 42(6): e410-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24633188

RESUMO

OBJECTIVES: Despite advances in cardiac arrest treatment, high mortality and morbidity rates after successful cardiopulmonary resuscitation are still a major clinical relevant problem. The post cardiac arrest syndrome subsumes myocardial dysfunction, impaired microcirculation, systemic inflammatory response, and neurological impairment. The calcium-sensitizer levosimendan was able to improve myocardial function and initial resuscitation success after experimental cardiac arrest/cardiopulmonary resuscitation. We hypothesized that levosimendan exerts beneficial effects on cerebral blood flow, neuronal injury, neurological outcome, and inflammation 24 hours after experimental cardiac arrest/cardiopulmonary resuscitation. DESIGN: Laboratory animal study. SETTING: University animal research laboratory. SUBJECTS: Sixty-one male Sprague-Dawley rats. INTERVENTIONS: Animals underwent asphyxial cardiac arrest/cardiopulmonary resuscitation, randomized to groups with levosimendan treatment (bolus 12 µg/kg and infusion for 3 hr [0.3 µg/min/kg]) or vehicle (saline 0.9% bolus and infusion for 3 hr [equivalent fluid volume]). Cardiac index, local cerebral blood flow, and hemodynamic variables were measured for 180 minutes after cardiac arrest/cardiopulmonary resuscitation. Behavioral and neurological evaluations were conducted 24 hours after cardiac arrest/cardiopulmonary resuscitation. Furthermore, neuronal injury, expressed as Fluoro-Jade B-positive cells in the hippocampal formation, cortical and hippocampal inflammatory cytokine gene expression, and blood plasma interleukin-6 values were assessed. MEASUREMENTS AND MAIN RESULTS: Treatment with levosimendan reduced neuronal injury and improved neurological outcome after 24 hours of reperfusion and resulted in elevated cardiac index and local cerebral blood flow compared with vehicle after cardiac arrest/cardiopulmonary resuscitation. Mean arterial blood pressure was reduced during the early reperfusion period in the levosimendan group. Cortical and hippocampal inflammatory cytokine gene expression and blood plasma interleukin-6 levels were not influenced. CONCLUSIONS: Levosimendan increased cerebral blood flow after experimental cardiac arrest/cardiopulmonary resuscitation. This effect coincided with reduced neuronal injury and improved neurologic outcome. Findings seem to be independent of inflammatory effects because no effects by levosimendan on cerebral or systemic inflammation could be detected. In summary, levosimendan is a promising agent to improve neurological outcome after cardiac arrest/cardiopulmonary resuscitation.


Assuntos
Reanimação Cardiopulmonar/efeitos adversos , Cardiotônicos/farmacologia , Circulação Cerebrovascular/efeitos dos fármacos , Parada Cardíaca/tratamento farmacológico , Hemodinâmica/efeitos dos fármacos , Hidrazonas/farmacologia , Neurônios/patologia , Piridazinas/farmacologia , Análise de Variância , Animais , Córtex Cerebral/metabolismo , Ensaio de Imunoadsorção Enzimática , Expressão Gênica , Parada Cardíaca/metabolismo , Hipocampo/metabolismo , Interleucina-6/sangue , Interleucina-6/genética , Masculino , Neurônios/citologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Simendana
2.
J Anat ; 224(4): 377-91, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24387791

RESUMO

The granulocyte colony-stimulating factor (G-CSF), being a member of the hematopoietic growth factor family, is also critically involved in controlling proliferation and differentiation of neural stem cells. Treatment with G-CSF has been shown to result in substantial neuroprotective and neuroregenerative effects in various experimental models of acute and chronic diseases of the central nervous system. Although G-CSF has been tested in a clinical study for treatment of acute ischemic stroke, there is only fragmentary data on the distribution of this cytokine and its receptor in the human brain. Therefore, the present study was focused on the immunohistochemical analysis of the protein expression of G-CSF and its receptor (G-CSF R) in the adult human brain. Since G-CSF has been shown not only to exert neuroprotective effects in animal models of Alzheimer's disease (AD) but also to be a candidate for clinical treatment, we have also placed an emphasis on the regulation of these molecules in this neurodegenerative disease. One major finding is that both G-CSF and G-CSF R were ubiquitously but not uniformly expressed in neurons throughout the CNS. Protein expression of G-CSF and G-CSF R was not restricted to neurons but was also detectable in astrocytes, ependymal cells, and choroid plexus cells. However, the distribution of G-CSF and G-CSF R did not substantially differ between AD brains and control, even in the hippocampus, where early neurodegenerative changes typically occur.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Fator Estimulador de Colônias de Granulócitos/metabolismo , Receptores de Fator Estimulador de Colônias de Granulócitos/metabolismo , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Neurônios/metabolismo
3.
Stroke ; 43(1): 185-92, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22020031

RESUMO

BACKGROUND AND PURPOSE: Both application of granulocyte-colony stimulating factor (G-CSF) and constraint-induced movement therapy (CIMT) have been shown to improve outcome after experimental stroke. The aim of the present study was to determine whether concurrent or sequential combination of both therapies will further enhance therapeutic benefit and whether specific modifications in the abundance of various neurotransmitter receptors do occur. METHODS: Adult male Wistar rats were subjected to photothrombotic ischemia and assigned to the following treatment groups (n=20 each): (1) ischemic control (saline); (2) CIMT (CIMT between poststroke Days 2 and 11; (3) G-CSF (10 µg/kg G-CSF daily between poststroke Days 2 and 11; (4) combined concurrent group (CIMT plus 10 µg/kg G-CSF daily between poststroke Days 2 and 11; and (5) combined sequential group (CIMT between poststroke Days 2 and 11 and 10 µg/kg G-CSF daily between poststroke Days 12 and 21, respectively). Rats were functionally tested before and up to 4 weeks after ischemia. Quantitative receptor autography was performed for N-methyl-d-aspartate, AMPA, and GABA(A) receptors. RESULTS: Significant improvement of functional outcome was seen in all groups treated with G-CSF alone and in either combination with CIMT, whereas CIMT alone failed to enhance recovery. Infarct sizes and remaining cortical tissue did not differ in the various treatment groups. Failure of significant benefit in the CIMT group was associated with a shift toward inhibition in perilesional and remote cortical regions. CONCLUSIONS: Our findings disclose G-CSF as the major player for enhanced recovery after experimental stroke, preventing a shift toward inhibition as seen in the CIMT group.


Assuntos
Isquemia Encefálica/terapia , Encéfalo/fisiologia , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Modalidades de Fisioterapia , Recuperação de Função Fisiológica/efeitos dos fármacos , Acidente Vascular Cerebral/terapia , Animais , Encéfalo/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/fisiopatologia , Terapia Combinada , Fator Estimulador de Colônias de Granulócitos/farmacologia , Masculino , Ratos , Ratos Wistar , Regeneração , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/fisiopatologia
4.
J Neural Transm (Vienna) ; 119(11): 1389-406, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22430742

RESUMO

Granulocyte-monocyte colony-stimulating factor (GM-CSF) is a member of the hematopoietic growth factor family, promoting proliferation and differentiation of hematopoietic progenitor cells of the myeloid lineage. In recent years, GM-CSF has also proved to be an important neurotrophic factor in the central nervous system (CNS) via binding to the GM-CSF receptor (GM-CSF R). Furthermore, studies on rodent CNS revealed a wide distribution of both the major binding α-subunit of the GM-CSF R (GM-CSF Rα) and its ligand. Since respective data on the expression pattern of these two molecules are still lacking, the present study has been designed to systematically analyze the protein expression of GM-CSF and GM-CSF Rα in the human brain, with particular emphasis on their regulation in Alzheimer's disease (AD). One major finding is that both GM-CSF and GM-CSF Rα were ubiquitously but not uniformly expressed in neurons throughout the CNS. Protein expression of GM-CSF and GM-CSF Rα was not restricted to neurons but also detectable in astrocytes, ependymal cells and choroid plexus cells. Interestingly, distribution and intensity of immunohistochemical staining for GM-CSF did not differ among AD brains and age-matched controls. Concerning GM-CSF Rα, a marked reduction of protein expression was predominantly detected in the hippocampus although a slight reduction was also found in various cortical regions, thalamic nuclei and some brainstem nuclei. Since the hippocampus is one of the target regions of neurodegenerative changes in AD, reduction of GM-CSF Rα, with consecutive downregulation of GM-CSF signaling, may contribute to in the progressive course of neurodegeneration in AD.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Regulação para Baixo/fisiologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/metabolismo , Neurônios/patologia , Subunidades Proteicas/metabolismo
5.
Brain ; 134(Pt 3): 704-20, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21354973

RESUMO

T lymphocytes are increasingly recognized as key modulators of detrimental inflammatory cascades in acute ischaemic stroke, but the potential of T cell-targeted therapy in brain ischaemia is largely unexplored. Here, we characterize the effect of inhibiting leukocyte very late antigen-4 and endothelial vascular cell adhesion molecule-1-mediated brain invasion-currently the most effective strategy in primary neuroinflammatory brain disease in murine ischaemic stroke models. Very late antigen-4 blockade by monoclonal antibodies improved outcome in models of moderate stroke lesions by inhibiting cerebral leukocyte invasion and neurotoxic cytokine production without increasing the susceptibility to bacterial infections. Gene silencing of the endothelial very late antigen-4 counterpart vascular cell adhesion molecule-1 by in vivo small interfering RNA injection resulted in an equally potent reduction of infarct volume and post-ischaemic neuroinflammation. Furthermore, very late antigen-4-inhibition effectively reduced the post-ischaemic vascular cell adhesion molecule-1 upregulation, suggesting an additional cross-signalling between invading leukocytes and the cerebral endothelium. Dissecting the specific impact of leukocyte subpopulations showed that invading T cells, via their humoral secretion (interferon-γ) and immediate cytotoxic mechanisms (perforin), were the principal pathways for delayed post-ischaemic tissue injury. Thus, targeting T lymphocyte-migration represents a promising therapeutic approach for ischaemic stroke.


Assuntos
Encéfalo/metabolismo , Movimento Celular/fisiologia , Encefalite/etiologia , Encefalite/patologia , Linfócitos/fisiologia , Acidente Vascular Cerebral/complicações , Animais , Anticorpos Monoclonais/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proteínas de Ligação a DNA/deficiência , Modelos Animais de Doenças , Encefalite/genética , Encefalite/prevenção & controle , Ensaio de Imunoadsorção Enzimática/métodos , Citometria de Fluxo , Transtornos Neurológicos da Marcha/etiologia , Integrina alfa4/imunologia , Integrina alfa4/metabolismo , Interferon gama/farmacologia , Leucócitos/efeitos dos fármacos , Leucócitos/fisiologia , Linfócitos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Perforina , Proteínas Citotóxicas Formadoras de Poros/deficiência , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/genética , Regulação para Cima/efeitos dos fármacos , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo
6.
Stroke ; 42(6): 1757-63, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21566228

RESUMO

BACKGROUND AND PURPOSE: Intravenous neural progenitor cell (NPC) treatment was shown to improve functional recovery after experimental stroke. The underlying mechanisms, however, are not completely understood so far. Here, we investigated the effects of systemic NPC transplantation on endogenous neurogenesis and dendritic plasticity of host neurons. METHODS: Twenty-four hours after photothrombotic ischemia, adult rats received either 5 million NPC or placebo intravenously. Behavioral tests were performed weekly up to 4 weeks after ischemia. Endogenous neurogenesis, dendritic length, and dendritic branching of cortical pyramid cells and microglial activation were quantified. RESULTS: NPC treatment led to a significantly improved sensorimotor function measured by the adhesive removal test. The dendritic length and the amount of branch points were significantly increased after NPC transplantation, whereas endogenous neurogenesis was decreased compared to placebo therapy. Decreased endogenous neurogenesis was associated with an increased number of activated microglial cells. CONCLUSIONS: Our findings suggest that an increased dendritic plasticity might be the structural basis of NPC-induced functional recovery. The decreased endogenous neurogenesis after NPC treatment seems to be mediated by microglial activation.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Células-Tronco Neurais/fisiologia , Células-Tronco Neurais/transplante , Neurogênese/fisiologia , Recuperação de Função Fisiológica/fisiologia , Acidente Vascular Cerebral/terapia , Animais , Comportamento Animal/fisiologia , Células Cultivadas , Camundongos , Células-Tronco Neurais/citologia , Fenótipo , Distribuição Aleatória , Ratos , Acidente Vascular Cerebral/fisiopatologia
7.
Neurocrit Care ; 15(3): 577-84, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21618067

RESUMO

BACKGROUND: Volatile anesthetics reduce postischemic neurohistopathological injury and improve neurological outcome in various animal models. However, the isoflurane concentrations above 1 minimum alveolar concentration (MAC) have been associated with reduced neuronal survival and impaired functional outcome. The aim of this study was to evaluate if 1.8 MAC sevoflurane alters postischemic neuronal survival and neurologic outcome compared with 0.45 MAC sevoflurane. METHODS: In this study, 20 fasted male Sprague-Dawley rats were randomly assigned to treatment groups with 1 or 4 vol.% sevoflurane end-tidal concentration. Cerebral ischemia was induced by bilateral carotid artery occlusion and hemorrhagic hypotension (BCAO). The cognitive outcome was assessed after 7 days using the object recognition test. Animals were then re-anesthetized and brains were removed for neurohistopathological analysis of the hippocampus (CA1) and cortex using hematoxylin-eosin staining. RESULTS: Physiologic parameters were not different between both the treatment groups. The number of viable neurons (median [Q1, Q3]) in the CA1 region on postischemic day 7 was increased after high-dose sevoflurane compared with low-dose sevoflurane (1645 [453, 1825] vs. 3222 [2920, 3993] neurons/ROI, P < 0.05). Results of the object recognition test were not different between both the treatment groups. CONCLUSIONS: Postischemic neuronal survival was increased with 1.8 MAC compared with 0.45 MAC sevoflurane. Therefore, experimental models of cerebral ischemia should account for neuroprotective effects of sevoflurane with increasing concentrations. To ensure minimal interference of sevoflurane on neuronal survival, a low inspired concentration should be used and fluctuations in the depth of anesthesia should be limited.


Assuntos
Anestésicos Inalatórios/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Comportamento Exploratório/efeitos dos fármacos , Ataque Isquêmico Transitório/patologia , Rememoração Mental/efeitos dos fármacos , Éteres Metílicos/farmacologia , Neurônios/efeitos dos fármacos , Reconhecimento Visual de Modelos/efeitos dos fármacos , Prosencéfalo/irrigação sanguínea , Prosencéfalo/efeitos dos fármacos , Animais , Discriminação Psicológica/efeitos dos fármacos , Relação Dose-Resposta a Droga , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Masculino , Exame Neurológico/efeitos dos fármacos , Neurônios/patologia , Prosencéfalo/patologia , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Sevoflurano
8.
PLoS One ; 11(1): e0146679, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26752421

RESUMO

We have previously shown that granulocyte-colony stimulating factor (G-CSF) treatment alone, or in combination with constraint movement therapy (CIMT) either sequentially or concomitantly, results in significantly improved sensorimotor recovery after photothrombotic stroke in rats in comparison to untreated control animals. CIMT alone did not result in any significant differences compared to the control group (Diederich et al., Stroke, 2012;43:185-192). Using a subset of rat brains from this former experiment the present study was designed to evaluate whether dendritic plasticity would parallel improved functional outcomes. Five treatment groups were analyzed (n = 6 each) (i) ischemic control (saline); (ii) CIMT (CIMT between post-stroke days 2 and 11); (iii) G-CSF (10 µg/kg G-CSF daily between post-stroke days 2 and 11); (iv) combined concurrent group (CIMT plus G-CSF) and (v) combined sequential group (CIMT between post-stroke days 2 and 11; 10 µg/kg G-CSF daily between post-stroke days 12 and 21, respectively). After impregnation of rat brains with a modified Golgi-Cox protocol layer V pyramidal neurons in the peri-infarct cortex as well as the corresponding contralateral cortex were analyzed. Surprisingly, animals with a similar degree of behavioral recovery exhibited quite different patterns of dendritic plasticity in both peri-lesional and contralesional areas. The cause for these patterns is not easily to explain but puts the simple assumption that increased dendritic complexity after stroke necessarily results in increased functional outcome into perspective.


Assuntos
Dendritos/patologia , Luz , Plasticidade Neuronal , Modalidades de Fisioterapia , Recuperação de Função Fisiológica , Acidente Vascular Cerebral/fisiopatologia , Acidente Vascular Cerebral/terapia , Animais , Terapia Combinada , Dendritos/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Masculino , Movimento , Plasticidade Neuronal/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Células Piramidais/patologia , Ratos Wistar , Recuperação de Função Fisiológica/efeitos dos fármacos
9.
Restor Neurol Neurosci ; 34(4): 665-75, 2016 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-26410211

RESUMO

PURPOSE: Using G-CSF deficient mice we recently demonstrated neuroprotective properties of endogenous G-CSF after ischemic stroke. The present follow-up study was designed to check, whether specific alterations in ligand binding densities of excitatory glutamate or inhibitory GABAA receptors may participate in this effect. METHODS: Three groups of female mice were subjected to 45 minutes of MCAO: wildtype, G-CSF deficient and G-CSF deficient mice substituted with G-CSF. Infarct volumes were determined after 24 hours and quantitative in vitro receptor autoradiography was performed using [3H]MK-801, [3H]AMPA and [3H]muscimol for labeling of NMDA, AMPA and GABAA receptors, respectively. Ligand binding densities were analyzed in regions in the ischemic core, peri-infarct areas and corresponding contralateral regions. RESULTS: Infarct volumes did not significantly differ between the experimental groups. Ligand binding densities of NMDA and GABAA receptors were widely in the same range. However, AMPA receptor binding densities in G-CSF deficient mice were substantially enhanced compared to wildtype mice. G-CSF substitution in mice lacking G-CSF largely reversed this effect. CONCLUSIONS: Although infarct volumes did not differ 24 hours after ischemia the increase of AMPA receptor binding densities in G-CSF deficient mice may explain the bigger infarcts previously observed at later time-points with the same stroke model.


Assuntos
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Fator Estimulador de Colônias de Granulócitos/metabolismo , Neuroproteção/fisiologia , Receptores de AMPA/metabolismo , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Animais , Autorradiografia , Feminino , Fator Estimulador de Colônias de Granulócitos/deficiência , Camundongos , Receptores de GABA-A/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
10.
Brain Pathol ; 23(1): 34-44, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22775530

RESUMO

Cellular and humoral inflammations play important roles in ischemic brain injury. The effectiveness of immunomodulatory therapies may critically depend on the chosen experimental model. Our purpose was to compare the post-ischemic neuroinflammation among murine permanent and transient middle cerebral artery occlusion (MCAO) models. Permanent MCAO was induced by transtemporal electrocoagulation and 30 minutes or 90 minutes transient MCAO was induced by intraluminal filament in C57BL/6 mice. Infiltration of leukocyte subpopulations was quantified by immunohistochemistry and fluorescence-activated cell sorting. Cerebral cytokine and adhesion molecule expression was measured by real-time polymerase chain reaction (RT-PCR). Neutrophil infiltration was noted at 24 h after transient MCAO, but did not further increase until 5 days in the permanent MCAO model. Few T cells were observed in both MCAO models at 24 h, but permanent MCAO demonstrated much more infiltrating T cells at 5 days. Pronounced microglial activation was evident at 24 h and 5 days after permanent but not after transient MCAO. The number of invading NK cells and expression of MHCII on CD11b+ cells did not differ among the three groups. Five days after MCAO, the expression of IL-1, TNF-α and IFN-γ and of the adhesion molecules ICAM-1 and VCAM-1 was significantly higher in the permanent than in the transient MCAO groups. Cellular and humoral inflammation differs substantially among commonly used MCAO models. Neuroinflammation is more pronounced after permanent electrocoagulatory MCAO compared with 30 minutes and 90 minutes filament-MCAO.


Assuntos
Encéfalo/fisiopatologia , Regulação Leucêmica da Expressão Gênica/fisiologia , Infarto da Artéria Cerebral Média/imunologia , Infarto da Artéria Cerebral Média/patologia , Infiltração de Neutrófilos/fisiologia , Análise de Variância , Animais , Antígenos CD/metabolismo , Antígenos Ly/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Moléculas de Adesão Celular/metabolismo , Córtex Cerebral/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/patologia , Subfamília B de Receptores Semelhantes a Lectina de Células NK/metabolismo , Linfócitos T/fisiologia , Fatores de Tempo
11.
PLoS One ; 6(6): e21312, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21701599

RESUMO

BACKGROUND: The contribution of neuroinflammation and specifically brain lymphocyte invasion is increasingly recognised as a substantial pathophysiological mechanism after stroke. FTY720 is a potent treatment for primary neuroinflammatory diseases by inhibiting lymphocyte circulation and brain immigration. Previous studies using transient focal ischemia models showed a protective effect of FTY720 but did only partially characterize the involved pathways. We tested the neuroprotective properties of FTY720 in permanent and transient cortical ischemia and analyzed the underlying neuroimmunological mechanisms. METHODOLOGY/PRINCIPAL FINDINGS: FTY720 treatment resulted in substantial reduction of circulating lymphocytes while blood monocyte counts were significantly increased. The number of histologically and flow cytometrically analyzed brain invading T- and B lymphocytes was significantly reduced in FTY720 treated mice. However, despite testing a variety of treatment protocols, infarct volume and behavioural dysfunction were not reduced 7d after permanent occlusion of the distal middle cerebral artery (MCAO). Additionally, we did not measure a significant reduction in infarct volume at 24 h after 60 min filament-induced MCAO, and did not see differences in brain edema between PBS and FTY720 treatment. Analysis of brain cytokine expression revealed complex effects of FTY720 on postischemic neuroinflammation comprising a substantial reduction of delayed proinflammatory cytokine expression at 3d but an early increase of IL-1ß and IFN-γ at 24 h after MCAO. Also, serum cytokine levels of IL-6 and TNF-α were increased in FTY720 treated animals compared to controls. CONCLUSIONS/SIGNIFICANCE: In the present study we were able to detect a reduction of lymphocyte brain invasion by FTY720 but could not achieve a significant reduction of infarct volumes and behavioural dysfunction. This lack of neuroprotection despite effective lymphopenia might be attributed to a divergent impact of FTY720 on cytokine expression and possible activation of innate immune cells after brain ischemia.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/imunologia , Imunossupressores/uso terapêutico , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Propilenoglicóis/uso terapêutico , Esfingosina/análogos & derivados , Animais , Edema Encefálico/tratamento farmacológico , Edema Encefálico/imunologia , Edema Encefálico/metabolismo , Isquemia Encefálica/metabolismo , Cloridrato de Fingolimode , Interleucina-6/metabolismo , Leucócitos/efeitos dos fármacos , Leucócitos/imunologia , Leucócitos/metabolismo , Linfócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Esfingosina/uso terapêutico , Fator de Necrose Tumoral alfa/metabolismo
12.
J Neuroimmunol ; 227(1-2): 1-9, 2010 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-20557950

RESUMO

Using a transgenic mouse model of ischemic stroke we checked for a possible interaction of antiphospholipid antibodies (aPL) which often cause thromboses as well as central nervous system (CNS) involvement under non-thrombotic conditions and the TWEAK/Fn14 pathway known to be adversely involved in inflammatory and ischemic brain disease. After 7 days, infarct volumes were reduced in Fn14 deficient mice and were further decreased by aPL treatment. This was associated with strongest increase of the endogenous neuroprotective G-CSF/G-CSF receptor system. This unexpected beneficial action of aPL is an example for a non-thrombogenic action and the double-edged nature of aPL.


Assuntos
Anticorpos Antifosfolipídeos/uso terapêutico , Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevenção & controle , Modelos Animais de Doenças , Fator Estimulador de Colônias de Granulócitos/biossíntese , Receptores do Fator de Necrose Tumoral/deficiência , Animais , Anticorpos Antifosfolipídeos/efeitos adversos , Isquemia Encefálica/patologia , Fator Estimulador de Colônias de Granulócitos/fisiologia , Humanos , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Distribuição Aleatória , Receptores de Fator Estimulador de Colônias de Granulócitos/biossíntese , Receptores de Fator Estimulador de Colônias de Granulócitos/fisiologia , Receptores do Fator de Necrose Tumoral/fisiologia , Transdução de Sinais/genética , Receptor de TWEAK
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA