Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Neurochem Int ; 108: 238-245, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28414094

RESUMEN

Aging is the greatest single risk factor of the neurodegenerative disorder Alzheimer's disease (AD). The monoaminergic system, including serotonin (5-HT), dopamine (DA) and noradrenaline (NA) modulates cognition, which is affected in AD. Changes in monoamine levels have been observed in AD, but these can both be age- and/or disease-related. We examined whether brain monoamine levels change as part of physiological aging and/or AD-like disease in APPSWE/PS1ΔE9 (APP/PS1) transgenic mice. The neocortex, hippocampus, striatum, brainstem and cerebellum of 6-, 12-, 18- and 24-month-old B6C3 wild-type (WT) mice and of 18-month old APP/PS1 and WT mice were analysed for 5-HT, DA and NA contents by high pressure liquid chromatography (HPLC), along with neocortex from 14-month-old APP/PS1 and WT mice. While, we observed no aging effect in WT mice, we detected region-specific changes in the levels of all monoamines in 18-month-old transgenic compared with WT mice. This included reductions in 5-HT (-30%), DA (-47%) and NA (-32%) levels in the neocortex and increases of 5-HT in the brainstem (+18%). No changes were observed in any of the monoamines in the neocortex from 14-month-old APP/PS1 mice. In combination, these findings indicate that aging alone is not sufficient to affect brain monoamine levels, unlike the APPSWE/PS1ΔE9 genotype.


Asunto(s)
Envejecimiento/metabolismo , Enfermedad de Alzheimer/metabolismo , Monoaminas Biogénicas/metabolismo , Encéfalo/metabolismo , Envejecimiento/genética , Envejecimiento/patología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/patología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Presenilina-1/genética
3.
Exp Brain Res ; 194(1): 17-27, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19139873

RESUMEN

Endonuclease G (EndoG) is a mitochondrial enzyme, known to be involved in caspase-independent cell death following translocation to the cellular nucleus. Nuclear translocation of EndoG has been observed in the ischemic area following transient occlusion of the middle cerebral artery (MCA) in mice, but not after permanent MCA occlusion. In this study we investigated the cellular and temporal expression of EndoG in infarcted cortex during the first 24 h after permanent MCA occlusion in mice, using immunohistochemistry, quantitative rt-PCR and cell specific immunoflourescence markers. EndoG translocated from the cytoplasm to the nucleus as early as 4 h and with a significant increase in the number of EndoG positive nuclei at 12 and 24 h after MCA occlusion. Nuclear translocation of EndoG was observed in degenerating NeuN positive neurons that were evenly distributed throughout the developing infarct. Translocation of EndoG was supported by unaltered EndoG mRNA levels. EndoG was neither expressed in GFAP positive astrocytes nor in CD11b positive microglia/macrophages. In contrast, CD11b positive microglia, but not infiltrating CD11b positive bone marrow-derived macrophages, were shown to express activated caspase-3. The translocation of EndoG to the nucleus of neurons in the infarct implicates EndoG in ischemic neuronal degeneration after permanent MCA occlusion in mice. Increased knowledge about EndoG involvement in ischemic neuronal cell death in mice might offer a promise to control processes involved in neuronal cell death pathways in stroke.


Asunto(s)
Corteza Cerebral/metabolismo , Endodesoxirribonucleasas/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Degeneración Nerviosa/metabolismo , Neuronas/metabolismo , Animales , Astrocitos/metabolismo , Antígeno CD11b/metabolismo , Caspasa 3/metabolismo , Corteza Cerebral/patología , Quimera , Proteínas de Unión al ADN , Técnica del Anticuerpo Fluorescente , Proteína Ácida Fibrilar de la Glía/metabolismo , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/patología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/ultraestructura , Proteínas Nucleares/metabolismo , Reacción en Cadena de la Polimerasa , ARN Mensajero
4.
Neuroscience ; 149(1): 112-22, 2007 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-17870248

RESUMEN

Microglia are innate immune cells and form the first line of defense of the CNS. Proliferation is a key event in the activation of microglia in acute pathology, and has been extensively characterized in rats, but not in mice. In this study we investigated axonal-lesion-induced microglial proliferation and surface antigen expression in C57BL/6 mice. Transection of the entorhino-dentate perforant path projection results in an anterograde axonal and a dense terminal degeneration that induces a region-specific activation of microglia in the dentate gyrus. Time-course analysis showed activation of microglial cells within the first week post-lesion and cell counting demonstrated a significant 1.6-fold increase in microglial numbers 24 h post-lesion reaching a maximal 3.8-fold increase 3 days post-lesion compared with controls. Double staining for the microglial macrophage antigen-1 and the proliferation marker bromodeoxyuridine, injected 1 h prior to perfusion, showed that lesion-reactive microglia accounted for the vast majority of proliferating cells. Microglia proliferated as soon as 24 h after lesion and 25% of all microglial cells were proliferating 3 days post-lesion. Immunofluorescence double staining showed that most activated, proliferating microglia occurred in multicellular clusters and co-expressed the intercellular adhesion molecule-1 and the hematopoietic stem cell marker cluster of differentiation 34. In conclusion, this study extends observations of axonal lesion-induced microglial proliferation in rats to mice, and provides new information on early microglial proliferation and microglial cluster formation and surface antigen expression in the mouse.


Asunto(s)
Axones/patología , Proliferación Celular , Regulación de la Expresión Génica/fisiología , Microglía/fisiología , Vía Perforante/lesiones , Animales , Antígenos CD34/metabolismo , Bromodesoxiuridina/metabolismo , Proteínas de Unión al Calcio/metabolismo , Recuento de Células/métodos , Fluoresceínas , Molécula 1 de Adhesión Intercelular/metabolismo , Antígeno de Macrófago-1/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Compuestos Orgánicos , Vía Perforante/patología , Factores de Tiempo
5.
Neuroscience ; 144(3): 934-49, 2007 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-17161916

RESUMEN

The proinflammatory and potential neurotoxic cytokine tumor necrosis factor (TNF) is produced by activated CNS resident microglia and infiltrating blood-borne macrophages in infarct and peri-infarct areas following induction of focal cerebral ischemia. Here, we investigated the expression of the TNF receptors, TNF-p55R and TNF-p75R, from 1 to 10 days following permanent occlusion of the middle cerebral artery in mice. Using quantitative polymerase chain reaction (PCR), we observed that the relative level of TNF-p55R mRNA was significantly increased at 1-2 days and TNF-p75R mRNA was significantly increased at 1-10 days following arterial occlusion, reaching peak values at 5 days, when microglial-macrophage CD11b mRNA expression was also increased. In comparison, the relative level of TNF mRNA was significantly increased from 1 to 5 days, with peak levels 1 day after arterial occlusion. In situ hybridization revealed mRNA expression of both receptors in predominantly microglial- and macrophage-like cells in the peri-infarct and subsequently in the infarct, and being most marked from 1 to 5 days. Using green fluorescent protein-bone marrow chimeric mice, we confirmed that TNF-p75R was expressed in resident microglia and blood-borne macrophages located in the peri-infarct and infarct 1 and 5 days after arterial occlusion, which was supported by Western blotting. The data show that increased expression of the TNF-p75 receptor following induction of focal cerebral ischemia in mice can be attributed to expression in activated microglial cells and blood-borne macrophages.


Asunto(s)
Infarto Encefálico/metabolismo , Gliosis/metabolismo , Macrófagos/metabolismo , Microglía/metabolismo , Receptores de Factor de Crecimiento Nervioso/genética , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Encéfalo/fisiopatología , Infarto Encefálico/fisiopatología , Antígenos CD11/genética , Citocinas/metabolismo , Gliosis/etiología , Gliosis/fisiopatología , Proteínas Fluorescentes Verdes , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Arteria Cerebral Media/patología , Arteria Cerebral Media/fisiopatología , ARN Mensajero/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Transducción de Señal/fisiología , Quimera por Trasplante , Receptores Señuelo del Factor de Necrosis Tumoral/genética , Regulación hacia Arriba/fisiología
6.
Neuroscience ; 132(4): 879-92, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15857694

RESUMEN

Interleukin-1beta (IL-1beta) is known to play a central role in ischemia-induced brain damage in rodents. In comparison to the rat, however, the available data on the cellular synthesis of IL-1beta mRNA and protein in the mouse are very limited. Here, we report on the time profile, the topography and the quantitative, cellular expression of IL-1beta mRNA in mice subjected to permanent occlusion of the distal middle cerebral artery (MCA). The in situ hybridization analysis showed that IL-1beta mRNA was expressed during the first post-surgical hour in a small number of high-expressing macrophage-like cells, located in cortical layers I and II of the future infarct. At 2 h, a significant number of faintly labeled IL-1beta mRNA-expressing cells had appeared in the developing peri-infarct, and the number remained constant at 4 h and 6 h, when the hybridization signal began to distribute to the cellular processes. Quantitative PCR performed on whole hemispheres showed a significant 20-fold increase in the relative level of IL-1beta mRNA at 12 h and a highly significant 42-fold increase at 24 h, at which time single IL-1beta mRNA-expressing cells were supplemented by aggregates and perivascular infiltrates of intensely labeled IL-1beta mRNA-expressing cells. Immunohistochemistry and double immunohistochemical stainings in addition to combined in situ hybridization, confirmed that the intensely labeled IL-1beta mRNA-expressing and IL-1beta protein synthesizing cells predominantly were glial fibrillary acidic protein-immunonegative, macrophage associated antigen-1-immunopositive microglia-macrophages. By day 5 there was a dramatic decline in the relative level of IL-1beta mRNA in the ischemic hemisphere. In summary, the data provide evidence that permanent occlusion of the distal MCA in mice results in expression of IL-1beta mRNA and IL-1beta synthesis in spatially and temporally segregated subpopulations of microglia and macrophages.


Asunto(s)
Encéfalo/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Interleucina-1/biosíntesis , Macrófagos/metabolismo , Microglía/metabolismo , Animales , Western Blotting , Encéfalo/patología , Inmunohistoquímica , Hibridación in Situ , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratones , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
7.
Lab Anim ; 38(4): 413-7, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15479556

RESUMEN

We adapted a non-invasive, fast, reliable and inexpensive procedure for the sampling and extraction of deoxyribonucleic acid (DNA) for genetic testing of mice. The procedure is based on a simple DNA extraction procedure used in the forensic genetic testing of humans. It involves mouth swabbing of the inner cheek using a cotton stick, followed by alkaline lysis of the harvested buccal epithelial cells. This procedure allows for repeated sampling and genetic testing of the individual mouse, and it is faster, simpler and, in our hands, more reliable than the currently used routine procedures for the sampling and extraction of mouse DNA. Current procedures all involve biopsy of a piece of the tail, ear or toe, followed by lengthy procedures to release and isolate the DNA.


Asunto(s)
ADN/aislamiento & purificación , Ratones/genética , Reacción en Cadena de la Polimerasa/veterinaria , Saliva/química , Manejo de Especímenes/veterinaria , Animales , Mejilla/fisiología , ADN/química , ADN/genética , Células Epiteliales/química , Reacción en Cadena de la Polimerasa/métodos , Manejo de Especímenes/métodos
10.
Exp Brain Res ; 138(3): 384-92, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11460777

RESUMEN

Although oligodendrocytes are vulnerable to focal cerebral ischemia, remyelination of denuded or regenerating axons in the peri-infarct area has been observed in the central nervous system. We studied the expression of myelin basic protein (MBP), a major component of central nervous system myelin, in peri-infarct areas in adult rat brain after transient middle cerebral artery occlusion (MCAO) and correlated it to the expression of the growth-associated protein-43 (GAP-43), a marker for axonal regeneration and sprouting, using non-radioactive in situ hybridization techniques. Within the infarct, MBP messenger RNA (mRNA) had disappeared by 24 h, whereas myelin protein, identified by MBP and myelin oligodendrocyte glycoprotein (MOG) immunohistochemistry, appeared structurally intact until day 3. Peri-infarct oligodendrocytes increased their expression of MBP mRNA from 24 h to maximal levels at day 7, corresponding to the appearance of process-bearing MBP and occasional MOG-immunoreactive oligodendrocytes in parallel sections. Quantitative analysis revealed significant increases in the density of oligodendrocytes (up to 7.6-fold) and in the level of MBP mRNA expressed by individual cells. Parallel sections showed that increased expression of GAP-43 mRNA in neurons was concomitant to MBP mRNA upregulation in oligodendrocytes. While the mechanisms regulating oligodendrocyte survival and myelination signals are not clear at this point, axonal sprouting could putatively serve as a stimulus for the upregulation of oligodendrocyte cell numbers, differentiation state, and/or active myelination in the peri-infarct areas.


Asunto(s)
Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Infarto Cerebral/metabolismo , Proteína GAP-43/metabolismo , Proteína Básica de Mielina/metabolismo , ARN Mensajero/metabolismo , Regulación hacia Arriba/genética , Animales , Encéfalo/patología , Encéfalo/fisiopatología , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Infarto Cerebral/patología , Infarto Cerebral/fisiopatología , Proteína GAP-43/genética , Regulación de la Expresión Génica/fisiología , Inmunohistoquímica , Hibridación in Situ , Masculino , Proteína Básica de Mielina/genética , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Ratas , Ratas Endogámicas SHR , Transcripción Genética/fisiología
11.
Brain Res Brain Res Protoc ; 7(2): 175-91, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11356385

RESUMEN

We present here sensitive, simple and robust methods for detection of tumor necrosis factor (TNF) mRNA and TNF in histological sections and homogenates of brain tissue from mice subjected to focal cerebral ischemia or hippocampal axonal lesioning. Both types of lesions are characterized by induction of TNF synthesis in resident microglial cells, which in the ischemic lesions are supplemented by TNF synthesizing, blood-borne macrophages. In situ hybridization for TNF mRNA is performed using alkaline phosphatase-labelled oligodeoxynucleotide probes. These probes show excellent rendition of individual cells, and can successfully be combined with immunohistochemical procedures. We also describe a sensitive immunohistochemical method for detection of TNF, which can be combined with visualization of an additional antigen. The specificity of the histological procedures are confirmed by RT-PCR and Western blot analysis on homogenates prepared from microdissected brain regions. Advantages and disadvantages of the methods are discussed with emphasis on the specificity and sensitivity of the histological procedures. Our strategy for detection of TNF mRNA and protein provides a solid basis for clarifying the cellular synthesis, regulation and function of TNF in the normal, injured or diseased CNS. Furthermore, the methodology can readily be applied in studies of other cytokines and growth factors in the CNS.


Asunto(s)
Corteza Cerebral/química , Inmunohistoquímica/métodos , Hibridación in Situ/métodos , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/genética , Animales , Western Blotting , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Cartilla de ADN , Femenino , Proteína Ácida Fibrilar de la Glía/análisis , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Antígeno de Macrófago-1/análisis , Masculino , Ratones , Ratones Endogámicos , Microglía/química , Vía Perforante/patología , Vía Perforante/fisiopatología , Vía Perforante/cirugía , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad
12.
Novartis Found Symp ; 231: 166-77; discussion 177-83, 302-6, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11131537

RESUMEN

Restorative transplantation of human embryonic nigral tissue for Parkinson's disease has given encouraging results with functional benefit and minimal signs of rejection in patients receiving standard immunosuppression. Due to the limited availability of human donor material and ethical concerns with its use, porcine tissue is considered an appropriate alternative. In animal studies, neural allo- and xenografts are usually rejected in the brain, emphasizing the necessity of understanding factors underlying survival and rejection of intracerebral neural transplants. Here, we review fundamental mechanisms of allo- and xenograft rejection, and discuss the privileged immune status of the brain, and how we may take advantage of this in order to improve and secure graft survival. Rejection of neural xenografts is expected to be of a cellular nature, like neural allograft rejection, but may also display unique features, and cannot be dealt with using conventional immunosuppressive therapies. The challenge therefore is to improve existing and design new strategies that allow permanent survival of histoincompatible neural grafts, taking advantage of the special immune status of adult CNS and immature donor brain tissue.


Asunto(s)
Trasplante de Tejido Encefálico/inmunología , Rechazo de Injerto/inmunología , Neuronas/citología , Neuronas/trasplante , Inmunología del Trasplante/inmunología , Animales , Rechazo de Injerto/patología , Humanos
13.
J Neurosci ; 20(10): 3612-21, 2000 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-10804203

RESUMEN

Glial reactivity is implicated in CNS repair and regenerative responses. Microglia, the cells responding earliest to axonal injury, produce tumor necrosis factor-alpha (TNFalpha), a cytokine with both cytopathic and neuroprotective effects. We have studied activation of hippocampal microglia to produce TNFalpha in response to transection of perforant path axons in SJL/J mice. TNFalpha mRNA was produced in a transient manner, peaking at 2 d and falling again by 5 d after lesioning. This was unlike other markers of glial reactivity, such as Mac-1 upregulation, which were sustained over longer time periods. Message for the immune cytokine interferon-gamma (IFNgamma) was undetectable, and glial reactivity to axonal lesions occurred as normal in IFNgamma-deficient mice. Microglial responses to lesion-induced neuronal injury were markedly enhanced in myelin basic protein promoter-driven transgenic mice, in which IFNgamma was endogenously produced in hippocampus. The kinetics of TNFalpha downregulation 5 d after lesion was not affected by transgenic IFNgamma, indicating that IFNgamma acts as an amplifier and not an inducer of response. These results are discussed in the context of a regenerative role for TNFalpha in the CNS, which is innately regulated and potentiated by IFNgamma.


Asunto(s)
Antineoplásicos/farmacología , Axones/patología , Hipocampo/patología , Interferón gamma/farmacología , Microglía/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética , Animales , Desnervación , Expresión Génica/inmunología , Hibridación in Situ , Antígeno de Macrófago-1/análisis , Antígeno de Macrófago-1/inmunología , Ratones , Ratones Transgénicos , Microglía/inmunología , Microglía/patología , Proteína Básica de Mielina/genética , Degeneración Nerviosa/inmunología , Degeneración Nerviosa/patología , Oligodendroglía/fisiología , Vía Perforante/patología , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/inmunología
14.
Int J Dev Neurosci ; 18(2-3): 221-35, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10715577

RESUMEN

The regulation of oligodendrocyte gene expression and myelination in vivo in the normal and injured adult CNS is still poorly understood. We have analyzed the effects of axotomy-induced axonal sprouting and microglial activation, on oligodendrocyte myelin basic protein (MBP) gene expression from 2 to 35 days after transection of the entorhino-hippocampal perforant path axonal projection. In situ hybridization analysis showed that anterograde axonal and terminal degeneration lead to upregulated oligodendrocyte MBP mRNA expression starting between day 2 and day 4, in (1) the deep part of stratum radiatum of CA3 and the dentate hilus, which display axonal sprouting but no degenerative changes or microglial activation, and (2) the outer part of the molecular layer of the fascia dentata, and in stratum moleculare of CA3 and stratum lacunosum-moleculare of CA1, areas that display dense anterograde axonal and terminal degeneration, myelin degenerative changes, microglial activation and axotomi-induced axonal sprouting. Oligodendrocyte MBP mRNA expression reached maximum in both these areas at day 7. MBP gene transcription remained constant in stratum radiatum, stratum pyramidale and stratum oriens of CA1, areas that were unaffected by perforant path transection. These results provide strong evidence that oligodendrocyte MBP gene expression can be regulated by axonal sprouting independently of microglial activation in the injured adult CNS.


Asunto(s)
Axones/enzimología , Hipocampo/fisiología , Proteína Básica de Mielina/genética , Regeneración Nerviosa/fisiología , Degeneración Walleriana/fisiopatología , Acetilcolinesterasa/análisis , Animales , Axones/química , Colorantes , Desnervación , Corteza Entorrinal/química , Corteza Entorrinal/citología , Corteza Entorrinal/fisiología , Expresión Génica/fisiología , Hipocampo/química , Hipocampo/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/fisiología , Plasticidad Neuronal/fisiología , Oligodendroglía/fisiología , Vía Perforante/química , Vía Perforante/citología , Vía Perforante/fisiología , ARN Mensajero/análisis , Tinción con Nitrato de Plata , Cloruro de Tolonio
15.
Brain Res ; 859(2): 394-7, 2000 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-10719094

RESUMEN

Neuroleptic-induced vacuous chewing movements (VCM) in rats, a putative analogue to tardive dyskinesia in man, may involve degeneration within striatum as well as changes in neurotransmitter and receptor expression. In this study, we measured the expression of dopamine D(1) receptor mRNA by dorsal striatal neurons in rats with high and low level of VCM after treatment with haloperidol for 38 weeks. Both the average integrated density of the in situ hybridization signal and number of cells obtained by the stereological cell counting remained within control level, irrespective of the level of haloperidol-induced oral dyskinesia.


Asunto(s)
Antagonistas de Dopamina/efectos adversos , Esquema de Medicación , Discinesia Inducida por Medicamentos/tratamiento farmacológico , Discinesia Inducida por Medicamentos/fisiopatología , Regulación de la Expresión Génica/efectos de los fármacos , Haloperidol/efectos adversos , Masticación/efectos de los fármacos , Masticación/fisiología , Neostriado/efectos de los fármacos , Neostriado/fisiopatología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Receptores de Dopamina D1/efectos de los fármacos , Receptores de Dopamina D1/genética , Animales , Recuento de Células , Femenino , Neostriado/citología , Neuronas/citología , Ratas , Ratas Sprague-Dawley
16.
Neurosci Lett ; 279(1): 21-4, 2000 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-10670778

RESUMEN

Neuroleptic-induced oral dyskinesia in rats, a putative analogue to human tardive dyskinesia, may be due to degeneration within the striatum. Using unbiased stereological methods, a decreased number of striatal neurons expressing preprosomatostatin mRNA was observed only in rats that developed pronounced oral dyskinesias after 30 weeks of haloperidol administration. The amount of preprosomatostatin mRNA in each striatal neuron, measured in terms of optical densities of individual neurons, was not affected by haloperidol. A tendency toward a reduction in the number of NADPH-diaphorase positive neurons was observed in rats receiving haloperidol. These results indicate that the mechanism by which neuroleptics induce oral dyskinesias in rats, and perhaps tardive dyskinesia in humans, involves a functional disruption and possibly damage of a subpopulation of interneurons in the striatum.


Asunto(s)
Discinesia Inducida por Medicamentos/patología , Haloperidol/efectos adversos , Neostriado/efectos de los fármacos , Neuronas/efectos de los fármacos , Precursores de Proteínas/genética , ARN Mensajero/análisis , Somatostatina/genética , Animales , Discinesia Inducida por Medicamentos/metabolismo , Femenino , Haloperidol/administración & dosificación , Neostriado/patología , Neuronas/patología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
17.
J Cereb Blood Flow Metab ; 20(1): 53-65, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10616793

RESUMEN

The proinflammatory cytokine tumor necrosis factor (TNF) is known to be expressed in brain ischemia; however, its cellular and temporal appearance is not fully settled. In this study, nonradioactive in situ hybridization for murine TNF mRNA was performed on brain sections from adult C57x129 mice at 6 hours, 12 hours, 24 hours, 2 days, 5 days, or 10 days (six to eight mice per group) after induction of permanent focal cerebral ischemia. Cortical infarct volumes were estimated, and TNF mRNA-expressing cells were counted within the infarct and infarct border using Cast-Grid analysis. At 12 hours, a peak of 19.2 +/- 5.1 TNF mRNA-expressing cells/mm2 was counted, contrasting two to three times lower values at 6 and 24 hours (6.4 +/- 4.6 and 9.2 +/- 3.4 cells/mm2, respectively) and <2 cells/mm2 at 48 hours and later stages. The TNF mRNA-expressing cells were distributed along the entire rostrocaudal axis of the cortical infarcts and occasionally within the caudate putamen. At all time points, TNF mRNA colocalized with Mac-1-positive microglia/macrophages but not with Ly-6G (Gr-1)-positive polymorphonuclear leukocytes. Similarly, combined in situ hybridization for TNF mRNA and immunohistochemistry for glial fibrillary acidic protein at 12 and 24 hours revealed no TNF mRNA-expressing astrocytes at these time points. Translation of TNF mRNA into bioactive protein was demonstrated in the neocortex of C57B1/6 mice subjected to permanent middle cerebral artery occlusion. In summary, this study points to a time-restricted microglial/macrophage production of TNF in focal cerebral ischemia in mice.


Asunto(s)
Arteriopatías Oclusivas/metabolismo , Arterias Cerebrales , Macrófagos/metabolismo , Microglía/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Arteriopatías Oclusivas/complicaciones , Isquemia Encefálica/complicaciones , Infarto Cerebral/etiología , Infarto Cerebral/metabolismo , Infarto Cerebral/patología , Antígeno de Macrófago-1/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/patología , ARN Mensajero/metabolismo , Factores de Tiempo , Distribución Tisular , Factor de Necrosis Tumoral alfa/genética
18.
Exp Brain Res ; 135(3): 319-30, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11146810

RESUMEN

Heparin affin regulatory peptide (HARP), also known as pleiotrophin or heparin-binding growth-associated molecule, is a developmentally regulated extracellular matrix protein that induces cell proliferation and promotes neurite outgrowth in vitro as well as pre- and postsynaptic developmental differentiation in vivo. Here we have investigated the expression of HARP mRNA and protein in the perforant path lesioned C57B1/6 mouse hippocampal formation from 1 to 35 days after surgery. This type of lesion induces a dense anterograde and terminal axonal degeneration, activation of glial cells, and reactive axonal sprouting within the perforant path zones of the fascia dentata and hippocampus as well as axotomy-induced retrograde neuronal degeneration in the entorhinal cortex. Analysis of sham- and unoperated control mice showed that HARP mRNA is expressed in neurons and white and gray matter glial cells as well as vascular and pial cells throughout the normal, adult brain. Lesioning induced high levels of HARP mRNA in astroglial-like cells in the denervated zones of fascia dentata and hippocampus as soon as day 2 postlesion. This expression reached maximum at day 4, and declined toward normal at day 7-14. Combined HARP in situ hybridization and glial fibrillary acidic protein (GFAP) immunohistochemical staining and double immunofluorescent stainings for GFAP and HARP at day 4 postlesion showed colocalization of HARP mRNA and protein to hypertrophic GFAP-immunopositive astrocytes in the denervated areas. Finally, the axotomized entorhinal layer II neurons, which expressed high levels of HARP mRNA in the normal brain, exhibited a marked decline in hybridization signal after axotomy. The induction of high levels of HARP mRNA and protein in astrocytes in the denervated areas of fascia dentata and hippocampus is of particular interest as astrocytes and astrocyte-derived factors are known to be implicated in axonal growth and regeneration and in rescuing injured neurons.


Asunto(s)
Proteínas Portadoras/biosíntesis , Citocinas/biosíntesis , Hipocampo/metabolismo , Factores de Crecimiento Nervioso/biosíntesis , Vía Perforante/fisiología , Animales , Colorantes , Colorantes Fluorescentes , Proteína Ácida Fibrilar de la Glía/metabolismo , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Degeneración Nerviosa , Proteínas del Tejido Nervioso/biosíntesis , Adhesión en Parafina , ARN Mensajero/biosíntesis
19.
Cell Transplant ; 8(5): 489-99, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10580343

RESUMEN

Metabolically compromised cells may be subject to complement-mediated cytotoxicity. The aim of this study was to clarify to what extent plasma complement C3 might contribute to the low survival (5-20%) of grafted dopaminergic neurons. The survival of intrastriatal cell suspension grafts of syngeneic dopaminergic, tyrosine hydroxylase (TH)-containing neurons was compared in rats subjected to short-term i.v. treatment with 1) cobra venom factor (CVF), or 2) placebo treatment. Depletion of plasma complement C3 by CVF was confirmed by crossed immunoelectrophoresis. With 159 +/- 37 (mean +/- SEM) TH-immunoreactive and 154 + /- 40 TH mRNA-expressing neurons in the CVF-treated rats (n = 9), and 117 +/- 34 TH-immunoreactive and 160 +/- 49 TH mRNA-expressing neurons in placebo rats (n = 6), the CVF treatment did not increase the survival of the grafted dopaminergic neurons. Similarly, CVF had no apparent effect on the astroglial, microglial, or oligodendroglial cell response within and around the graft. The data indicate that depletion of plasma complement C3 at the time of grafting has no effect on the long-term survival of syngeneic ventral mesencephalic dopaminergic neuronal grafts.


Asunto(s)
Antígenos CD , Antígenos de Neoplasias , Proteínas Aviares , Proteínas Sanguíneas , Trasplante de Tejido Encefálico/fisiología , Complemento C3/antagonistas & inhibidores , Cuerpo Estriado , Venenos Elapídicos/farmacología , Trasplante de Tejido Fetal/fisiología , Supervivencia de Injerto/efectos de los fármacos , Neuronas/trasplante , Animales , Antígenos de Superficie/análisis , Basigina , Trasplante de Tejido Encefálico/métodos , Cuerpo Estriado/citología , Trasplante de Tejido Fetal/métodos , Feto , Edad Gestacional , Proteína Ácida Fibrilar de la Glía/análisis , Antígeno de Macrófago-1/análisis , Masculino , Glicoproteínas de Membrana/análisis , Neuronas/citología , Neuronas/fisiología , Ratas , Ratas Endogámicas WKY , Transcripción Genética , Trasplante Isogénico , Tirosina 3-Monooxigenasa/análisis , Tirosina 3-Monooxigenasa/genética
20.
Neuroscience ; 93(2): 507-18, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10465434

RESUMEN

Transection of the entorhino-dentate perforant path is a well known model for lesion-induced axonal sprouting and glial reactions in the rat. In this study, we have characterized the microglial reaction in the dentate molecular layer of the SJL/J and C57Bl/6 mouse. The morphological transformation of the microglial cells and their densitometrically measured Mac-1 immunoreactivity were correlated with the density of silver-impregnated axonal and terminal degeneration and the myelination of the degenerating medial and lateral perforant pathways. Anterograde axonal and terminal degeneration leads to: (i) altered myelin basic protein immunoreactivity with the appearance of discrete myelin deposits preferentially in the denervated medial and significantly less so in the lateral perforant path zone from day 2 after lesioning; (ii) an increase in number and Mac-1 immunoreactivity of morphologically-changed microglial cells in the denervated perforant path zones with more pronounced morphological transformation of microglia in the medial than in the lateral perforant path zones at day 2 but not day 5 after lesioning; and (iii) a linear correlation between the density of microglial Mac-1 reactivity and axonal degeneration in the medial but not in the lateral perforant path zone at two days postlesion, and a linear correlation in both zones at five days postlesion. We propose that the differentiated microglial response is due to the different densities of axonal and terminal degeneration, as observed in the individual cases. The finding of a potentiated or accelerated microglial activation in the medial as compared to the lateral perforant path zone suggests different kinetics of microglial activation in areas with degenerating myelinated and unmyelinated fibers.


Asunto(s)
Axones/fisiología , Giro Dentado/fisiología , Microglía/fisiología , Vaina de Mielina/fisiología , Degeneración Nerviosa/patología , Vía Perforante/fisiología , Terminales Presinápticos/fisiología , Animales , Colorantes , Densitometría , Giro Dentado/citología , Giro Dentado/ultraestructura , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Proteína Básica de Mielina/metabolismo , Oligodendroglía/metabolismo , Tinción con Nitrato de Plata , Cloruro de Tolonio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...