Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Exp Neurol ; 271: 155-67, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26024859

RESUMEN

Although a physiological function of the cellular prion protein (PrP(c)) is still not fully clarified, a PrP(c)-mediated neuroprotection against hypoxic/ischemic insult is intriguing. After ischemic stroke prion protein knockout mice (Prnp(0/0)) display significantly greater lesions as compared to wild-type (WT) mice. Earlier reports suggested an interaction between the glycolytic enzyme lactate dehydrogenase (LDH) and PrP(c). Since hypoxic environment enhances LDH expression levels and compels neurons to rely on lactate as an additional oxidative substrate for energy metabolism, we examined possible differences in LDH protein expression in WT and Prnp(0/0) knockout models under normoxic/hypoxic conditions in vitro and in vivo, as well as in a HEK293 cell line. While no differences are observed under normoxic conditions, LDH expression is markedly increased after 60-min and 90-min of hypoxia in WT vs. Prnp(0/0) primary cortical neurons with concurrent less hypoxia-induced damage in the former group. Likewise, cerebral ischemia significantly increases LDH levels in WT vs. Prnp(0/0) mice with accompanying smaller lesions in the WT group. HEK293 cells overexpressing PrP(c) show significantly higher LDH expression/activity following 90-min of hypoxia as compared to control cells. Moreover, a cytoplasmic co-localization of LDH and PrP(c) was recorded under both normoxic and hypoxic conditions. Interestingly, an expression of monocarboxylate transporter 1, responsible for cellular lactate uptake, increases with PrP(c)-overexpression under normoxic conditions. Our data suggest LDH as a direct PrP(c) interactor with possible physiological relevance under low oxygen conditions.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Hipoxia/genética , Hipoxia/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Priones/metabolismo , Animales , Infarto Encefálico/etiología , Infarto Encefálico/metabolismo , Hipoxia de la Célula/fisiología , Modelos Animales de Enfermedad , Electroforesis en Gel Bidimensional , Embrión de Mamíferos , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Hipoxia/complicaciones , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transportadores de Ácidos Monocarboxílicos/metabolismo , Neuronas/metabolismo , Proteínas Priónicas , Priones/genética , Factores de Tiempo , Transfección , Tubulina (Proteína)/metabolismo
2.
Stem Cells ; 30(6): 1297-310, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22593021

RESUMEN

Novel therapeutic concepts against cerebral ischemia focus on cell-based therapies in order to overcome some of the side effects of thrombolytic therapy. However, cell-based therapies are hampered because of restricted understanding regarding optimal cell transplantation routes and due to low survival rates of grafted cells. We therefore transplanted adult green fluorescence protein positive neural precursor cells (NPCs) either intravenously (systemic) or intrastriatally (intracerebrally) 6 hours after stroke in mice. To enhance survival of NPCs, cells were in vitro protein-transduced with TAT-heat shock protein 70 (Hsp70) before transplantation followed by a systematic analysis of brain injury and underlying mechanisms depending on cell delivery routes. Transduction of NPCs with TAT-Hsp70 resulted in increased intracerebral numbers of grafted NPCs after intracerebral but not after systemic transplantation. Whereas systemic delivery of either native or transduced NPCs yielded sustained neuroprotection and induced neurological recovery, only TAT-Hsp70-transduced NPCs prevented secondary neuronal degeneration after intracerebral delivery that was associated with enhanced functional outcome. Furthermore, intracerebral transplantation of TAT-Hsp70-transduced NPCs enhanced postischemic neurogenesis and induced sustained high levels of brain-derived neurotrophic factor, glial cell line-derived neurotrophic factor, and vascular endothelial growth factor in vivo. Neuroprotection after intracerebral cell delivery correlated with the amount of surviving NPCs. On the contrary, systemic delivery of NPCs mediated acute neuroprotection via stabilization of the blood-brain-barrier, concomitant with reduced activation of matrix metalloprotease 9 and decreased formation of reactive oxygen species. Our findings imply two different mechanisms of action of intracerebrally and systemically transplanted NPCs, indicating that systemic NPC delivery might be more feasible for translational stroke concepts, lacking a need of in vitro manipulation of NPCs to induce long-term neuroprotection.


Asunto(s)
Isquemia Encefálica/terapia , Proteínas HSP70 de Choque Térmico/genética , Células-Madre Neurales/fisiología , Células-Madre Neurales/trasplante , Accidente Cerebrovascular/terapia , Animales , Isquemia Encefálica/metabolismo , Isquemia Encefálica/cirugía , Diferenciación Celular/fisiología , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Proteínas HSP70 de Choque Térmico/biosíntesis , Proteínas HSP70 de Choque Térmico/metabolismo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Trasplante de Células Madre/métodos , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/cirugía , Transducción Genética
3.
Am J Pathol ; 178(6): 2823-31, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21641403

RESUMEN

Although the pathologic role of the prion protein in transmissible spongiform encephalopathic diseases has been widely investigated, the physiologic role of the cellular prion protein (PrP(C)) is not known. Among the many functions attributed to PrP(C), there is increasing evidence that it is involved in cell survival and mediates neuroprotection. A potential role in the immune response has also been suggested. However, how these two functions interplay in autoimmune disease is unclear. To address this, autoimmune optic neuritis, a model of multiple sclerosis, was induced in C57Bl/6 mice, and up-regulation of PrP(C) was observed throughout the disease course. In addition, compared with wild-type mice, in PrP(C)-deficient mice and mice overexpressing PrP(C), histopathologic analysis demonstrated that optic neuritis was exacerbated, as indicated by axonal degeneration, inflammatory infiltration, and demyelination. However, significant neuroprotection of retinal ganglion cells, the axons of which form the optic nerve, was observed in mice that overexpressed PrP(C). Conversely, mice lacking PrP(C) demonstrated significantly more neurodegeneration. This suggests that PrP(C) may have a neuroprotective function independent of its role in regulating the immune response.


Asunto(s)
Enfermedades Autoinmunes/complicaciones , Enfermedades Autoinmunes/patología , Fármacos Neuroprotectores/farmacología , Neuritis Óptica/complicaciones , Neuritis Óptica/patología , Priones/metabolismo , Animales , Axones/efectos de los fármacos , Axones/patología , Citoprotección/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/complicaciones , Encefalomielitis Autoinmune Experimental/patología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Degeneración Nerviosa/complicaciones , Degeneración Nerviosa/patología , Nervio Óptico/efectos de los fármacos , Nervio Óptico/patología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Regulación hacia Arriba/efectos de los fármacos
4.
Neurobiol Dis ; 40(1): 265-76, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20554038

RESUMEN

Neural precursor cells (NPC) are an interesting tool in experimental stroke research, but their therapeutic potential is limited due to poor long-term survival. We therefore in vitro transduced subventricular zone-(SVZ)-derived NPC with the anti-apoptotic fusion protein TAT-Bcl-x(L) and analyzed NPC survival, differentiation, and post-stroke functional deficits after experimental ischemia in mice. Survival of TAT-Bcl-x(L)-transduced NPC, which were injected at day 7 post-stroke into the ischemic striatum, was significantly increased at 4 weeks after stroke. Increased survival of NPC was associated with reduced infarct injury and decreased post-stroke functional deficits. Animals grafted with TAT-Bcl-x(L)-transduced NPC showed an increased number of immature cells expressing the neuronal marker doublecortin. Since mature neuronal differentiation of NPC was not observed, reduced post-stroke injury cannot be attributed to enhanced neuronal regeneration, but rather to indirect by-stander effects of grafted NPC. In line with this, NPC-mediated neuroprotection of cortical neurons in vitro was associated with increased secretion of growth factors. Thus, in vitro transduction of cultivated NPC with TAT-Bcl-x(L) results in enhanced resistance of transplanted NPC followed by long-term neuroprotection and ameliorated functional deficits after transient focal cerebral ischemia in mice.


Asunto(s)
Isquemia Encefálica/genética , Isquemia Encefálica/terapia , Células-Madre Neurales/trasplante , Trasplante de Células Madre/métodos , Accidente Cerebrovascular/terapia , Transducción Genética/métodos , Proteína bcl-X/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Animales , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Isquemia Encefálica/cirugía , Supervivencia Celular/genética , Citoprotección/genética , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Recuperación de la Función/genética , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/patología , Tiempo , Proteína bcl-X/uso terapéutico , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/uso terapéutico
6.
Exp Neurol ; 223(2): 548-56, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20156439

RESUMEN

Endogenous neurogenesis persists in the subgranular zone (SGZ) of the adult rodent brain. Cerebral ischemia stimulates endogenous neurogenesis involving proliferation, migration and differentiation of SGZ-derived neural precursor cells (NPC). However, the biological meaning of this phenomenon is limited by poor survival of NPC. In order to study the effects of an acute neuroprotective treatment on hippocampal endogenous neurogenesis after transient cerebral ischemia in mice, we applied a fusion protein consisting of the TAT domain of the HI virus with the anti-apoptotic Bcl-x(L). Intravenous injection of TAT-Bcl-x(L) resulted in reduced hippocampal cell injury for up to 4weeks after stroke as assessed by TUNEL and NeuN staining. This was in line with a TAT-Bcl-x(L)-mediated reduced postischemic microglia activation. Analysis of endogenous hippocampal cell proliferation revealed an increased number of BrdU(+) cells in the TAT-Bcl-x(L) group 4weeks after stroke compared to animals treated with saline and TAT-HA (negative control). Cell proliferation in non-ischemic sham operated animals was not affected by TAT-Bcl-x(L). Twenty-eight days after stroke co-expression of BrdU(+) cells with the immature neuronal marker doublecortin was significantly increased in TAT-Bcl-x(L) animals. Although TAT-Bcl-x(L) treatment also resulted in an increased number of BrdU(+) cells expressing the mature neuronal marker NeuN, the total amount of these cells was low. These data show that TAT-Bcl-x(L) treatment yields both postischemic sustained hippocampal neuroprotection and increased survival of NPC rather than an induction of endogenous neurogenesis itself.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Hipocampo/citología , Neurogénesis/efectos de los fármacos , Proteína bcl-X/farmacología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología , Animales , Isquemia Encefálica/patología , Bromodesoxiuridina/metabolismo , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Encefalitis/tratamiento farmacológico , Encefalitis/patología , Hipocampo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Microglía/efectos de los fármacos , Neurogénesis/fisiología , Neuronas/citología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Proteínas Recombinantes de Fusión/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/patología
7.
J Cereb Blood Flow Metab ; 29(6): 1187-96, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19384335

RESUMEN

Cerebral ischemia stimulates endogenous neurogenesis within the subventricular zone and the hippocampal dentate gyrus of the adult rodent brain. However, such newly generated cells soon die after cerebral ischemia. To enhance postischemic survival of neural precursor cells (NPC) and long-lasting neural regeneration, we applied the antiapoptotic chaperone heat shock protein 70 (Hsp70) fused to a cell-penetrating peptide derived from the HIV TAT to ensure delivery across the blood-brain barrier and the cell membrane. After transient focal cerebral ischemia in mice, TAT-Hsp70 was intravenously injected concomitant with reperfusion and additionally on day 14 after stroke. TAT-Hsp70 treatment resulted in smaller infarct size (27.1+/-9.0 versus 109.0+/-14.0 and 88.5+/-26.0 mm(3) in controls) and in functional improvement as assessed by the rota rod, tight rope, and water maze tests when compared with saline- and TAT-hemagglutinin-treated controls. In addition, postischemic survival of endogenous doublecortin (Dcx)-positive NPC was improved within the lesioned striatum of TAT-Hsp70-treated animals for up to 4 weeks after stroke without changing overall cell proliferation of BrdU(+) cells. Thus, TAT-Hsp70 treatment after stroke may be a promising tool to act neuroprotective and improve postischemic functional outcome, and also to increase survival of endogenous NPC after stroke.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Productos del Gen tat/uso terapéutico , Proteínas HSP70 de Choque Térmico/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Animales , Bromodesoxiuridina/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteína Doblecortina , Productos del Gen tat/genética , Productos del Gen tat/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/metabolismo , Ratas
8.
Neurobiol Dis ; 34(1): 87-94, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19167500

RESUMEN

Cerebral ischemia activates endogenous neurogenesis in the subventricular zone (SVZ) and the dentate gyrus. Consecutively, SVZ-derived neural precursors migrate towards ischemic lesions. However, functional relevance of activated neurogenesis is limited by poor survival of new-born precursors. We therefore employed the HI-virus-derived fusion protein TAT-Bcl-x(L) to study the effects of acute anti-apoptotic treatment on endogenous neurogenesis and functional outcome after transient cerebral ischemia in mice. TAT-Bcl-x(L) treatment led to significantly reduced acute ischemic cell death (128+/-23 vs. 305+/-65 TUNEL+ cells/mm(2) in controls) and infarct volumes resulting in less motor deficits and improved spatial learning. It significantly increased survival of doublecortin (Dcx)-positive neuronal precursors (389+/-96 vs. 213+/-97 Dcx+ cells in controls) but did not enhance overall post-ischemic cell proliferation or lesion-specific neuronal differentiation 28 days after ischemia. Our data demonstrate that post-stroke TAT-Bcl-x(L)-treatment results in acute neuroprotection, improved functional outcome, and enhanced survival of lesion-specific neuronal precursor cells after cerebral ischemia in mice.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Cuerpo Estriado/fisiopatología , Productos del Gen tat/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Células Madre/fisiología , Proteína bcl-X/uso terapéutico , Animales , Isquemia Encefálica/fisiopatología , Muerte Celular/fisiología , Supervivencia Celular/fisiología , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Aprendizaje , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Actividad Motora , Neurogénesis/fisiología , Neuropéptidos/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Percepción Espacial
9.
Stroke ; 39(10): 2867-75, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18658037

RESUMEN

BACKGROUND AND PURPOSE: Systemic injection of hematopoietic stem cells after ischemic cardiac or neural lesions is one approach to promote tissue repair. However, mechanisms of possible protective or reparative effects are poorly understood. In this study we analyzed the effect of lineage-negative bone marrow-derived hematopoietic stem and precursor cells (Lin(-)-HSCs) on ischemic brain injury in mice. METHODS: Lin(-)-HSCs were injected intravenously at 24 hours after onset of a 45-minute transient cerebral ischemia. Effects of Lin(-)-HSCs injection on infarct size, apoptotic cell death, postischemic inflammation and cytokine gene transcription were analyzed. RESULTS: Green fluorescent protein (GFP)-marked Lin(-)-HSCs were detected at 24 hours after injection in the spleen and later in ischemic brain parenchyma, expressing microglial but no neural marker proteins. Tissue injury assessment showed significantly smaller infarct volumes and less apoptotic neuronal cell death in peri-infarct areas of Lin(-)-HSC-treated animals. Analysis of immune cell infiltration in ischemic hemispheres revealed a reduction of invading T cells and macrophages in treated mice. Moreover, Lin(-)-HSC therapy counter-regulated proinflammatory cytokine and chemokine receptor gene transcription within the spleen. CONCLUSIONS: Our data demonstrate that systemically applied Lin(-)-HSCs reduce cerebral postischemic inflammation, attenuate peripheral immune activation and mediate neuroprotection after ischemic stroke.


Asunto(s)
Isquemia Encefálica/patología , Isquemia Encefálica/terapia , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Inflamación/terapia , Animales , Apoptosis/fisiología , Isquemia Encefálica/inmunología , Movimiento Celular/inmunología , Citometría de Flujo , Células Madre Hematopoyéticas/metabolismo , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Inflamación/etiología , Inflamación/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/inmunología , Bazo/metabolismo
10.
Restor Neurol Neurosci ; 26(1): 57-64, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18431006

RESUMEN

PURPOSE: The physiological function of the cellular prion protein (PrPC) is still unclear. A growing body of evidence suggests that PrPC has neuroprotective properties and that its deletion increases susceptibility to focal cerebral ischemia. The purpose of this study was to elucidate the role of PrPC overexpression in ischemic brain injury in vivo. METHODS: PrPC overexpressing (TG35) and wild type (WT) mice were subjected to a 90-minute transient focal cerebral ischemia followed by infarct volume analysis 24 hours after lesion. To identify effects of PrPC overexpression on signalling pathways important for the regulation of ischemic cell death, we studied postischemic activation and expression of Akt and Erk1/2 using quantitative Western Blot analysis. RESULTS: TG35 mice displayed significantly smaller infarct volumes and showed reduced early postischemic Erk1/2 phosphorylation, a pathway known to exacerbate neuronal injury following transient cerebral ischemia. In contrast, PrPC overexpression did not change postischemic Akt phosphorylation, which acts anti-apoptotic and is reduced in PrPC knockout animals. CONCLUSIONS: These results demonstrate that PrPC overexpression reduces deleterious Erk1/2 activation but does not affect Akt activation after transient cerebral ischemia, suggesting a role for distinct cytosolic signalling pathways in PrPC mediated neuroprotection.


Asunto(s)
Isquemia Encefálica/prevención & control , Regulación de la Expresión Génica/fisiología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Oncogénica v-akt/metabolismo , Proteínas PrPC/metabolismo , Animales , Isquemia Encefálica/complicaciones , Infarto Cerebral/etiología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Transgénicos , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Modelos Biológicos , Fosforilación , Proteínas PrPC/genética , Factores de Tiempo
11.
J Neurosci Res ; 85(8): 1734-43, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17455321

RESUMEN

Recombinant tissue plasminogen activator (rt-PA) treatment improves functional outcome after acute ischemic stroke, inducing reperfusion by its thrombolytic activity. Conversely, there is evidence that rt-PA can mediate neuronal damage after ischemic brain injury in vivo. In addition to other mechanisms, enhancement of N-methyl-D-aspartate (NMDA) receptor signalling has been proposed to underlie rt-PA-mediated neurotoxicity. However, the role of poly(ADP-ribose) polymerase-1 (PARP-1) activation, which mediates postischemic excitotoxic cell death, in rt-PA-mediated aggravation of ischemic brain injury has not been established and was therefore addressed in this study. After permanent focal cerebral ischemia, intravenous rt-PA application significantly increased early postischemic PARP-1 activation within ischemic hemispheres and infarct volumes compared with control mice without affecting cerebral blood flow. Rt-PA induced increase in PARP-1 activation, and infarct volumes could be blocked by the PARP inhibitor 3-aminobenzamide. Moreover, the rt-PA-induced increase in PARP-1 activation was also prevented by the NMDA antagonist MK-801. In summary, we demonstrate that rt-PA treatment enhances postischemic PARP-1 activation, which contributes to rt-PA induced aggravation of ischemic brain injury in vivo. Furthermore, we provide evidence that NMDA receptor activation is required for rt-PA-mediated effects on postischemic PARP-1 activation.


Asunto(s)
Isquemia Encefálica/inducido químicamente , Poli(ADP-Ribosa) Polimerasas/metabolismo , Activador de Tejido Plasminógeno/efectos adversos , Animales , Benzamidas/farmacología , Análisis de los Gases de la Sangre , Presión Sanguínea/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/patología , Infarto Encefálico/inducido químicamente , Infarto Encefálico/enzimología , Infarto Encefálico/patología , Isquemia Encefálica/enzimología , Isquemia Encefálica/patología , Maleato de Dizocilpina/farmacología , Activación Enzimática , Ataque Isquémico Transitorio/inducido químicamente , Ataque Isquémico Transitorio/enzimología , Ataque Isquémico Transitorio/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Poli(ADP-Ribosa) Polimerasa-1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/fisiología , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/uso terapéutico , Activador de Tejido Plasminógeno/uso terapéutico
12.
Stroke ; 37(5): 1296-300, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16574930

RESUMEN

BACKGROUND AND PURPOSE: The physiological function of cellular prion protein (PrPc) is not yet understood. Recent findings suggest that PrPc may have neuroprotective properties, and its absence increases susceptibility to neuronal injury. The purpose of this study was to elucidate the role of PrPc in ischemic brain injury in vivo. METHODS: PrP knockout (Prnp(0/0)) and Prnp(+/+) wild-type (WT) mice were subjected to 60-minute transient or permanent focal cerebral ischemia followed by infarct volume analysis 24 hours after lesion. To identify effects of PrPc deletion on mechanisms regulating ischemic cell death, expression analysis of several proapoptotic and antiapoptotic proteins was performed at 6 and 24 hours after transient ischemia and in nonischemic controls using Western blot or immunohistochemistry. RESULTS: Prnp(0/0) mice displayed significantly increased infarct volumes after both transient or permanent ischemia when compared with WT animals (70.2+/-23 versus 13.3+/-4 mm3; 119.8+/-24 versus 86.4+/-25 mm3). Expression of phospho-Akt (Ser473) was significantly reduced in Prnp(0/0) compared with WT animals both early after ischemia and in sham controls. Furthermore, postischemic caspase-3 activation was significantly enhanced in the basal ganglia and the parietal cortex of Prnp(0/0) mice. In contrast, expression of total Akt, Bax, and Bcl-2 did not differ between both groups. CONCLUSIONS: These results demonstrate that PrPc deletion impairs the antiapoptotic phosphatidylinositol 3-kinase/Akt pathway by resulting in reduced postischemic phospho-Akt expression, followed by enhanced postischemic caspase-3 activation, and aggravated neuronal injury after transient and permanent cerebral ischemia.


Asunto(s)
Isquemia Encefálica/genética , Isquemia Encefálica/fisiopatología , Caspasas/metabolismo , Priones/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Animales , Isquemia Encefálica/metabolismo , Caspasa 3 , Caspasas/genética , Muerte Celular , Activación Enzimática/genética , Eliminación de Gen , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/fisiopatología , Ataque Isquémico Transitorio/genética , Ataque Isquémico Transitorio/metabolismo , Ataque Isquémico Transitorio/fisiopatología , Ratones , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal/genética
13.
Neurobiol Dis ; 18(3): 582-90, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15755684

RESUMEN

Pilocarpine-induced status epilepticus (PCSE) is a widely used model to study neurodegeneration in limbic structures after prolonged epileptic seizures. However, mechanisms mediating neuronal cell death in this model require further characterization. Examining the expression time course and spatial distribution of activated caspase-3, we sought to determine the role of apoptosis in PCSE-mediated neuronal cell death. Expression of activated caspase-3, predominantly located in neurons, was detected 24 h (amygdala; piriform and temporal cortex) and 7 days (hippocampus; amygdala; piriform, temporal and parietal cortex; thalamus) after PCSE with strongest induction being observed in the amygdala, the piriform cortex, and the hippocampus. Further analysis revealed TUNEL positivity (24 h and 7 days after SE) and a significant, progressive neuronal cell loss in all brain regions displaying caspase-3 activation. Corresponding to high levels of activated caspase-3 expression, neuronal cell loss was most pronounced in the amygdala, piriform cortex, and dorsal CA-1 hippocampus. These results demonstrate that apoptosis contributes significantly to PCSE-induced neuronal cell death.


Asunto(s)
Caspasas/biosíntesis , Regulación Enzimológica de la Expresión Génica/fisiología , Neuronas/enzimología , Convulsiones/enzimología , Estado Epiléptico/enzimología , Animales , Caspasa 3 , Caspasas/metabolismo , Muerte Celular/fisiología , Activación Enzimática/genética , Masculino , Neuronas/química , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
14.
Neurosci Lett ; 372(1-2): 146-50, 2004 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-15531106

RESUMEN

The pathological isoform of the prion protein (PrP(Sc)) has been identified to mediate transmissible spongiform encephalopathies like Creutzfeldt-Jakob disease (CJD). In contrast, the physiological function of the normal cellular prion protein (PrP(c)) is not yet understood. Recent findings suggest that PrP(c) may have neuroprotective properties and that its absence increases susceptibility to oxidative stress and neuronal injury. To determine whether PrP(c) is part of the cellular response to neuronal injury in vivo, we investigated PrP(c) regulation after severe and mild focal ischemic brain injury in mice using the thread occlusion stroke model. Western Blot and ELISA analysis showed a significant upregulation of PrP(c) in the ischemic hemisphere at 4 and 8h after onset of permanent focal ischemia, which was no longer detectable at 24h after lesion induction when compared to control animals. In contrast, transient focal ischemia (60 min) did only lead to slightly but not significantly elevated PrP(c) levels in the ischemic hemisphere when compared to controls. These results demonstrate that cerebral PrP(c) is upregulated early in response to focal cerebral ischemia. The extent of upregulation, however, seems to depend on the severity of ischemia and may therefore reflect the extent of ischemia induced neuronal damage. Given the known neuroprotective effects of PrP(c) in vitro, ischemia-induced upregulation of cerebral PrP(c) supports the hypothesis that, as part of an early adaptive cellular response to ischemic brain injury, PrP(c) may be involved in the regulation of ischemia-induced neuronal cell death in vivo.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Proteínas PrPC/biosíntesis , Regulación hacia Arriba/fisiología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL
15.
J Neurol ; 251(7): 838-44, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15258786

RESUMEN

Carotid endarterectomy (CEA) has been shown to be effective in stroke prevention in selected patients. Some studies, however, identified gender as an independent risk factor for perioperative CEA complications demonstrating an increased rate of perioperative stroke or death in women. Furthermore, contralateral internal carotid artery (ICA) occlusion has been associated with higher rates of perioperative CEA complications. Therefore, we sought to analyse the gender-specific risk of perioperative CEA complications between patients with or without contralateral ICA stenosis or occlusion. We retrospectively analysed 212 consecutive CEA patients (male = 156, Female = 56) for their gender-specific, perioperative risk of stroke, transient ischemic attack (TIA), restenosis and cardiac complications in the presence (62%) or absence (38%) of a contralateral ICA stenosis/occlusion. In women but not in men, risk of perioperative ischemic events (stroke, TIA) or re-stenosis (p = 0.036) and combined perioperative complications (ischemic events, re-stenosis or cardiac complications; 38.2 % vs. 9.1%; p = 0.028) was significantly increased in the presence of a contralateral ICA stenosis or occlusion. Furthermore, in the presence of a contralateral ICA stenosis/occlusion the number of perioperative ischemic events (p = 0.008) and combined perioperative complications (38.2 % vs. 14.3%; p = 0.006) was significantly higher in female than in male patients. Our study suggests that women with contralateral ICA stenosis or occlusion may have a significantly higher risk for perioperative CEA complications than other subgroups of CEA patients. This risk increase seems to be gender-specific and, if confirmed in larger prospective studies, may influence ICA stenosis therapy in the presence of a contralateral ICA stenosis/occlusion in female patients.


Asunto(s)
Estenosis Carotídea/cirugía , Endarterectomía Carotidea/efectos adversos , Complicaciones Intraoperatorias/epidemiología , Complicaciones Posoperatorias/epidemiología , Arteria Carótida Interna/cirugía , Estenosis Carotídea/patología , Comorbilidad , Femenino , Humanos , Ataque Isquémico Transitorio/epidemiología , Masculino , Isquemia Miocárdica/epidemiología , Recurrencia , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Distribución por Sexo , Factores Sexuales , Accidente Cerebrovascular/epidemiología
16.
Eur J Neurosci ; 19(2): 247-57, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14725618

RESUMEN

The establishment of retino-collicular topography is a well-investigated model of axon pathfinding and it was believed that this topography is irreversibly fixed throughout life. We now report that, after partial crush of the adult rat optic nerve, the anterograde transport of intravitreally-injected tracers via axons of surviving retinal ganglion cells (RGC) in all retinal quadrants is confined to the rostro-medial part of the superior colliculus (SC). This indicates that the retino-collicular topography is rearranged after partial crush of the adult rat optic nerve. The reorganization starts in the injured optic nerve where surviving axonal fibres are demyelinized and bundled in the periphery of the optic nerve distal to the crush site. This is followed by a displacement of surviving axons to the medial part of the optic tract (OT) within 2 weeks. The infiltration of macrophages with the subsequent production of tumour necrosis factor-alpha at the lesion site is a prerequisite for the altered retino-collicular projection as blockade of tumour necrosis factor-alpha signalling with the neutralizing antibody Infliximab abolishes reorganization in the SC and lateralization of RGC axons in the optic nerve and OT. This suggests that optic nerve inflammation is necessary for a progressive bundling of surviving RGC axons, probably via clearance of cellular debris which, in turn, may lead to a redistribution of RGC axons to the medial OT and rostro-medial SC.


Asunto(s)
Plasticidad Neuronal/fisiología , Traumatismos del Nervio Óptico/patología , Retina/fisiología , Colículos Superiores/fisiología , Animales , Vías Nerviosas/patología , Vías Nerviosas/fisiología , Ratas , Ratas Sprague-Dawley , Retina/patología , Colículos Superiores/patología
17.
J Neurol ; 249(1): 33-42, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11954866

RESUMEN

MRI including diffusion-weighted sequences (DW-MRI) has demonstrated its high sensitivity for acute supratentorial ischemic lesions. In this study we examined the sensitivity of different MRI sequences for the detection of acute brainstem and isolated thalamic infarctions. Diffusion- and T2-weighted MRI of 45 consecutive patients with signs and symptoms of infratentorial and thalamic infarction between 6/1997 and 1/2000 were analysed. The time between the onset of symptoms and the first MRI varied between 2 hours to 7 days with a median of 2 days. MRI repeats were performed in 4 patients in whom the clinical brainstem infarction had not been detected initially. Lesion detectability and size were evaluated for different brainstem and thalamic localizations. An acute brainstem or thalamic infarction as defined by the clinical condition could be identified in all patients by comparison of DW-MRI and T2-weighted images. Pons in farctions were the largest, followed by midbrain and thalamic lesions. Medulla oblongata infarctions were small in comparison. Pons, mid-brain and thalamic infarctions were reliably identified beginning 12 hours after the onset of symptoms. In contrast, detectability of medulla oblongata infarctions varied within the first 24 hours and their overall visibility was worse than that of other brainstem infarctions corresponding to their small size. However, regardless of loca tion, none of the 3 infarctions examined within the first 5 hours after the onset of symptoms could be identified. These lesions were demonstrated in follow-up examinations. In conclusion, pontine, midbrain and thalamic infarctions can reliably be visualized by a combination of DW-MRI and T2-weighted images beginning 12 hours after the ischemic attack. However, sensitivity seems to be lower earlier than 12 hours after ischemia and for medulla oblongata lesions.


Asunto(s)
Isquemia Encefálica/patología , Infartos del Tronco Encefálico/patología , Imagen por Resonancia Magnética/métodos , Enfermedades Talámicas/patología , Enfermedad Aguda , Adulto , Anciano , Anciano de 80 o más Años , Isquemia Encefálica/fisiopatología , Infartos del Tronco Encefálico/fisiopatología , Femenino , Humanos , Masculino , Bulbo Raquídeo/irrigación sanguínea , Bulbo Raquídeo/patología , Bulbo Raquídeo/fisiopatología , Mesencéfalo/irrigación sanguínea , Mesencéfalo/patología , Mesencéfalo/fisiopatología , Persona de Mediana Edad , Puente/irrigación sanguínea , Puente/patología , Puente/fisiopatología , Reproducibilidad de los Resultados , Estudios Retrospectivos , Estadística como Asunto , Enfermedades Talámicas/fisiopatología , Tálamo/irrigación sanguínea , Tálamo/patología , Tálamo/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...