Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Ther ; 32(3): 783-799, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38196192

RESUMEN

We recently described a novel ribosome-based regulatory mechanism/checkpoint that controls innate immune gene translation and microglial activation in non-sterile inflammation orchestrated by RNA binding protein SRSF3. Here we describe a role of SRSF3 in the regulation of microglia/macrophage activation phenotypes after experimental stroke. Using a model-system for analysis of the dynamic translational state of microglial ribosomes we show that 24 h after stroke highly upregulated immune mRNAs are not translated resulting in a marked dissociation of mRNA and protein networks in activated microglia/macrophages. Next, microglial activation after stroke was characterized by a robust increase in pSRSF3/SRSF3 expression levels. Targeted knockdown of SRSF3 using intranasal delivery of siRNA 24 h after stroke caused a marked knockdown of endogenous protein. Further analyses revealed that treatment with SRSF3-siRNA alleviated translational arrest of selected genes and induced a transient but significant increase in innate immune signaling and IBA1+ immunoreactivity peaking 5 days after initial injury. Importantly, delayed SRSF3-mediated increase in immune signaling markedly reduced the size of ischemic lesion measured 7 days after stroke. Together, our findings suggest that targeting SRSF3 and immune mRNA translation may open new avenues for molecular/therapeutic reprogramming of innate immune response after ischemic injury.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular , Humanos , Microglía/metabolismo , Isquemia Encefálica/genética , Isquemia Encefálica/terapia , Macrófagos/metabolismo , Accidente Cerebrovascular/patología , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo
2.
Neurotherapeutics ; 18(2): 1095-1112, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33786804

RESUMEN

Vascular dementia is one of the most common forms of dementia in aging population. However, the molecular mechanisms involved in development of disease and the link between the cerebrovascular pathology and the cognitive impairments remain elusive. Currently, one common and/or converging neuropathological pathway leading to dementia is the mislocalization and altered functionality of the TDP-43. We recently demonstrated that brain ischemia triggers an age-dependent deregulation of TDP-43 that was associated with exacerbated neurodegeneration. Here, we report that chronic cerebral hypoperfusion in mice (CCH) produced by unilateral common carotid artery occlusion induces cytoplasmic mislocalization of TDP-43 and formation of insoluble phosho-TDP-43 aggregates reminiscent of pathological changes detected in cortical neurons of human brain samples from patients suffering from vascular dementia. Moreover, the CCH in mice caused chronic activation of microglia and innate immune response, development of cognitive deficits, and motor impairments. Oral administration of a novel analog (IMS-088) of withaferin A, an antagonist of nuclear factor-κB essential modulator (NEMO), led to mitigation of TDP-43 pathology, enhancement of autophagy, and amelioration of cognitive/motor deficits in CCH mice. Taken together, our results suggest that targeting TDP-43 pathogenic inclusions may have a disease-modifying effect in dementia caused by chronic brain hypoperfusion.


Asunto(s)
Circulación Cerebrovascular/efectos de los fármacos , Trastornos Cerebrovasculares/genética , Disfunción Cognitiva/genética , Proteínas de Unión al ADN/genética , Trastornos Motores/genética , Proteinopatías TDP-43/genética , Animales , Circulación Cerebrovascular/fisiología , Trastornos Cerebrovasculares/tratamiento farmacológico , Trastornos Cerebrovasculares/patología , Enfermedad Crónica , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/patología , Sistemas de Liberación de Medicamentos/métodos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trastornos Motores/tratamiento farmacológico , Trastornos Motores/patología , Proteinopatías TDP-43/tratamiento farmacológico , Proteinopatías TDP-43/patología , Witanólidos/administración & dosificación , Witanólidos/química
3.
Brain Behav Immun Health ; 3: 100041, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34589834

RESUMEN

Growing evidence suggests that galectin-3 (Gal-3) is instrumental in orchestrating innate immune response and microglia activation following different brain pathologies. However, its role remains controversial. We recently showed that a readily available natural product glucosamine may act as a strong modulator of Gal-3. Glucosamine is a naturally occurring sugar and a precursor in the synthesis of glycosylated proteins. It is often used as a supplement to treat symptoms of various inflammatory conditions. Our recent work suggests that by increasing the synthesis and availability of Gal-3 ligands and/or by regulating its expression levels, glucosamine may significantly modulate Gal-3 signaling. Because evidence suggests that Gal-3 might be differentially regulated after ischemic injury in the brains of female mice, here we examined and compared the immunomodulatory potential of glucosamine in male and female stroke. The mice were subjected to transient middle cerebral artery occlusion (MCAO), followed by different reperfusion periods. The short-term 5 days treatment with glucosamine (150 â€‹mg/kg i.p.) was initiated 2 â€‹hrs after stroke. To visualize the effects of glucosamine treatment on post-stroke inflammation, we took advantage of a transgenic mouse model bearing the dual reporter system luciferase/GFP under transcriptional control of a murine TLR2 promoter (TLR2-luc-GFP) allowing in vivo bioluminescence imaging of innate immune response and microglial activation. We report that after stroke, both, male and female mice strongly up-regulate the TLR2 bioluminescence signals from activated microglia, however, the observed in vivo immunomodulatory effects of glucosamine after stroke were sex-dependent. Analysis of cytokine profiles at protein level, in glucosamine-treated male mice 72hsr after stroke, revealed down regulation of pro-inflammatory cytokines, an increase in levels of anti-inflammatory cytokines including IL-4, IL13 and colony stimulating factors MCFC and GM-CSF and a significant decrease in the size of ischemic lesion in male mice. Conversely, in female mice glucosamine markedly increases the pro-inflammatory signaling and exacerbates ischemic injury. Analysis of the downstream signaling target of glucosamine/Gal-3 revealed that glucosamine administration restored PPAR-γ activity in male but not in female mice 3 days following MCAO. Together, our results suggest that glucosamine acts as a fine tuner of post-ischemic inflammation in a sex dependent-manner and may have therapeutic potential after stroke in males. Based on our results propose that targeting immune system after stroke may require adapted sex-specific therapeutic approaches.

4.
J Neuroinflammation ; 15(1): 312, 2018 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-30413172

RESUMEN

BACKGROUND: TDP-43 has been identified as a disease-associated protein in several chronic neurodegenerative disorders and increasing evidence suggests its potentially pathogenic role following brain injuries. Normally expressed in nucleus, under pathological conditions TDP-43 forms cytoplasmic ubiquitinated inclusions in which it is abnormally phosphorylated and cleaved to generate a 35 and a 25 kDa C-terminal fragments. In the present study, we investigated age-related expression patterns of TDP-43 in neurons and glia and its role as modulator of inflammation following ischemic injury. METHODS: Wild-type and TDP-43 transgenic mice of different age groups were subjected to transient middle cerebral artery occlusion. The role of TDP-43 in modulation of inflammation was assessed using immunofluorescence, Western blot analysis, and in vivo bioluminescence imaging. Finally, post-mortem stroke human brain sections were analyzed for TDP-43 protein by immunohistochemistry. RESULTS: We report here an age-related increase and formation of ubiquitinated TDP-43 cytoplasmic inclusions after stroke. The observed deregulation in TDP-43 expression patterns was associated with an increase in microglial activation and innate immune signaling as revealed by in vivo bioluminescence imaging and immunofluorescence analysis. The presence of ubiquitinated TDP-43 aggregates and its cleaved TDP-35 and TDP-25 fragments was markedly increased in older, 12-month-old mice leading to larger infarctions and a significant increase in in neuronal death. Importantly, unlike the hallmark neuropathological features associated with chronic neurodegenerative disorders, the TDP-43-positive cytoplasmic inclusions detected after stroke were not phosphorylated. Next, we showed that an increase and/or overexpression of the cytoplasmic TDP-43 drives the pathogenic NF-κB response and further increases levels of pro-inflammatory markers and ischemic injury after stroke in age-dependent manner. Finally, analyses of the post-mortem stroke brain tissues revealed the presence of the cytoplasmic TDP-43 immunoreactive structures after human stroke. CONCLUSION: Together, our findings suggest that the level of cytoplasmic TDP-43 increases with aging and may act as an age-related mediator of inflammation and neuronal injury after stroke. Thus, targeting cytoplasmic TDP-43 may have a therapeutic potential after stroke.


Asunto(s)
Envejecimiento , Regulación de la Expresión Génica/fisiología , Inflamación/etiología , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/patología , Receptor Toll-Like 2/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Infarto Encefálico/etiología , Citocinas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Mutación/genética , Fosfopiruvato Hidratasa/metabolismo , Agregación Patológica de Proteínas/etiología , Agregación Patológica de Proteínas/genética , Receptor Toll-Like 2/genética
5.
J Neuroinflammation ; 14(1): 45, 2017 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-28253906

RESUMEN

BACKGROUND: Nestin is a known marker of neuronal progenitor cells in the adult brain. Following neuro- and gliogenesis, nestin is replaced by cell type-specific intermediate filaments, e.g., neurofilaments for panneuronal expression and glial fibrillary acidic protein as a specific marker of mature astrocytes. While previous work have been mostly focused on the neuronal fate of nestin-positive progenitors, in the present study, we sought to investigate in real time how nestin signals and cellular expression patterns are controlled in the context of neuroinflammatory challenge and ischemic brain injury. METHODS: To visualize effects of neuroinflammation on neurogenesis/gliogenesis, we created a transgenic model bearing the dual reporter system luciferase and GFP under transcriptional control of the murine nestin promoter. In this model, transcriptional activation of nestin was visualized from the brains of living animals using biophotonic/bioluminescence molecular imaging and a high resolution charged coupled device camera. Nestin induction profiles in vivo and in tissue sections were analyzed in two different experimental paradigms: middle cerebral artery occlusion and lipopolysaccharide-induced innate immune stimuli. RESULTS: We report here a context- and injury-dependent induction and cellular expression profile of nestin. While in the baseline conditions the nestin signal and/or GFP expression was restricted to neuronal progenitors, the cellular expression patterns of nestin following innate immune challenge and after stroke markedly differed shifting the cellular expression patterns towards activated microglia/macrophages and astrocytes. CONCLUSIONS: Our results suggest that nestin may serve as a context-dependent biomarker of inflammatory response in glial cells including activated microglia/macrophages.


Asunto(s)
Química Encefálica , Encéfalo/metabolismo , Mediadores de Inflamación/metabolismo , Microglía/metabolismo , Imagen Molecular/métodos , Nestina/metabolismo , Animales , Biomarcadores/metabolismo , Células Cultivadas , Inflamación/metabolismo , Mediadores de Inflamación/análisis , Mediciones Luminiscentes/métodos , Ratones , Ratones Transgénicos , Microglía/química , Nestina/análisis , Ratas
6.
Neurobiol Aging ; 40: 50-60, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26973103

RESUMEN

Estrogens are known to exert neuroprotective and immuneomodulatory effects after stroke. However, at present, little is known about the role of estrogens and its receptors in postischemic inflammation after menopause. Here, we provide important in vivo evidence of a distinct shift in microglial phenotypes in the model of postmenopause brain. Using a model-system for live imaging of microglial activation in the context of chronic estrogen- and ERα-deficiency associated with aging, we observed a marked deregulation of the TLR2 signals and/or microglial activation in ovariectomized and/or ERα knockout mice. Further analysis revealed a 5.7-fold increase in IL-6, a 4.7-fold increase in phospho-Stat3 levels suggesting an overactivation of JAK/STAT3 pathway and significantly larger infarction in ERα knockouts chronically deprived of estrogen. Taken together, our results suggest that in the experimental model of menopause and/or aging, ERα mediates innate immune responses and/or microglial activation, and ischemia-induced production of IL-6. Based on our results, we propose that the loss of functional ERα may lead to deregulation of postischemic inflammatory responses and increased vulnerability to ischemic injury in aging female brains.


Asunto(s)
Envejecimiento , Isquemia Encefálica/patología , Encéfalo/patología , Receptor alfa de Estrógeno/deficiencia , Receptor alfa de Estrógeno/fisiología , Menopausia , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Estrógenos/deficiencia , Estrógenos/fisiología , Femenino , Inmunidad Innata , Interleucina-6/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/patología , Modelos Animales , Factor de Transcripción STAT3/metabolismo
7.
J Neuroinflammation ; 9: 191, 2012 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-22873409

RESUMEN

BACKGROUND: Using a live imaging approach, we have previously shown that microglia activation after stroke is characterized by marked and long-term induction of the Toll-like receptor (TLR) 2 biophotonic signals. However, the role of TLR2 (and potentially other TLRs) beyond the acute innate immune response and as early neuroprotection against ischemic injury is not well understood. METHODS: TLR2-/- mice were subjected to transient middle cerebral artery occlusion followed by different reperfusion times. Analyses assessing microglial activation profile/innate immune response were performed using in situ hybridization, immunohistochemistry analysis, flow cytometry and inflammatory cytokine array. The effects of the TLR2 deficiency on the evolution of ischemic brain injury were analyzed using a cresyl violet staining of brain sections with appropriate lesion size estimation. RESULTS: Here we report that TLR2 deficiency markedly affects post-stroke immune response resulting in delayed exacerbation of the ischemic injury. The temporal analysis of the microglia/macrophage activation profiles in TLR2-/- mice and age-matched controls revealed reduced microglia/macrophage activation after stroke, reduced capacity of resident microglia to proliferate as well as decreased levels of monocyte chemotactic protein-1 (MCP-1) and consequently lower levels of CD45(high)/CD11b(+) expressing cells as shown by flow cytometry analysis. Importantly, although acute ischemic lesions (24 to 72 h) were smaller in TLR2-/- mice, the observed alterations in innate immune response were more pronounced at later time points (at day 7) after initial stroke, which finally resulted in delayed exacerbation of ischemic lesion leading to larger chronic infarctions as compared with wild-type mice. Moreover, our results revealed that TLR2 deficiency is associated with significant decrease in the levels of neurotrophic/anti-apoptotic factor Insulin-like growth factor-1 (IGF-1), expressed by microglia in the areas both in and around ischemic lesion. CONCLUSION: Our results clearly suggest that optimal and timely microglial activation/innate immune response is needed to limit neuronal damage after stroke.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Receptor Toll-Like 2/deficiencia , Animales , Isquemia Encefálica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Microglía/patología , Factores de Tiempo , Receptor Toll-Like 2/biosíntesis , Receptor Toll-Like 2/genética
8.
J Neurosci ; 32(30): 10383-95, 2012 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-22836271

RESUMEN

Growing evidence suggests that galectin-3 is involved in fine tuning of the inflammatory responses at the periphery, however, its role in injured brain is far less clear. Our previous work demonstrated upregulation and coexpression of galectin-3 and IGF-1 in a subset of activated/proliferating microglial cells after stroke. Here, we tested the hypothesis that galectin-3 plays a pivotal role in mediating injury-induced microglial activation and proliferation. By using a galectin-3 knock-out mouse (Gal-3KO), we demonstrated that targeted disruption of the galectin-3 gene significantly alters microglia activation and induces ∼4-fold decrease in microglia proliferation. Defective microglia activation/proliferation was further associated with significant increase in the size of ischemic lesion, ∼2-fold increase in the number of apoptotic neurons, and a marked deregulation of the IGF-1 levels. Next, our results revealed that contrary to WT cells, the Gal3-KO microglia failed to proliferate in response to IGF-1. Moreover, the IGF-1-mediated mitogenic microglia response was reduced by N-glycosylation inhibitor tunicamycine while coimmunoprecipitation experiments revealed galectin-3 binding to IGF-receptor 1 (R1), thus suggesting that interaction of galectin-3 with the N-linked glycans of receptors for growth factors is involved in IGF-R1 signaling. While the canonical IGF-1 signaling pathways were not affected, we observed an overexpression of IL-6 and SOCS3, suggesting an overactivation of JAK/STAT3, a shared signaling pathway for IGF-1/IL-6. Together, our findings suggest that galectin-3 is required for resident microglia activation and proliferation in response to ischemic injury.


Asunto(s)
Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Proliferación Celular/efectos de los fármacos , Galectina 3/metabolismo , Microglía/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Células Cultivadas , Galectina 3/genética , Factor I del Crecimiento Similar a la Insulina/farmacología , Interleucina-6/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/patología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
9.
Mol Imaging ; 10(6): 434-45, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22201534

RESUMEN

Although it has been well established that induction of growth-associated protein-43 (GAP-43) during development coincides with axonal outgrowth and early synapse formation, the existence of neuronal plasticity and neurite outgrowth in the adult central nervous system after injuries is more controversial. To visualize the processes of neuronal injury and repair in living animals, we generated reporter mice for bioluminescence and fluorescence imaging bearing the luc (luciferase) and gfp (green fluorescent protein) reporter genes under the control of the murine GAP-43 promoter. Reporter functionality was first observed during the development of transgenic embryos. Using in vivo bioluminescence and fluorescence imaging, we visualized induction of the GAP-43 signals from live embryos starting at E10.5, as well as neuronal responses to brain and peripheral nerve injuries (the signals peaked at 14 days postinjury). Moreover, three-dimensional analysis of the GAP-43 bioluminescent signal confirmed that it originated from brain structures affected by ischemic injury. The analysis of fluorescence signal at cellular level revealed colocalization between endogenous protein and the GAP-43-driven gfp transgene. Taken together, our results suggest that the GAP-43-luc/gfp reporter mouse represents a valid model system for real-time analysis of neurite outgrowth and the capacity of the adult nervous system to regenerate after injuries.


Asunto(s)
Lesiones Encefálicas/patología , Imagen Molecular/métodos , Neuronas/fisiología , Cicatrización de Heridas/fisiología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Química Encefálica , Lesiones Encefálicas/metabolismo , Trastornos Cerebrovasculares/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos , Femenino , Proteína GAP-43/biosíntesis , Proteína GAP-43/genética , Genes Reporteros , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Luciferasas/biosíntesis , Luciferasas/genética , Mediciones Luminiscentes/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuritas/metabolismo , Plasticidad Neuronal , Neuronas/metabolismo , Neuronas/patología , Regiones Promotoras Genéticas , Reproducibilidad de los Resultados , Espectrometría de Fluorescencia , Cicatrización de Heridas/genética
10.
Stroke ; 42(10): 2903-9, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21852616

RESUMEN

BACKGROUND AND PURPOSE: Consumption of fish has been shown to reduce risk of coronary heart disease and, possibly, of ischemic stroke. Because docosahexaenoic acid (DHA) is the most likely neuroactive component within fish oil, we hypothesized that exposing mice to a DHA-enriched diet may reduce inflammation and protect neurons against ischemic injury. METHODS: To visualize the effects of DHA on neuroinflammation after stroke, TLR2-fluc-GFP transgenic mice were exposed to either a control diet, a diet depleted in n-3 polyunsaturated fatty acid, or a diet enriched in DHA during 3 months. Real-time biophotonic/bioluminescence imaging of the TLR2 response was performed before and after middle cerebral artery occlusion, whereas cytokines concentrations and stroke area analyses were performed at 3 and 7 days after middle cerebral artery occlusion, respectively. RESULTS: We show that a 3-month DHA treatment prevented microglial activation after ischemic injury, reduced the ischemic lesion size, and increased levels of the antiapoptotic molecule Bcl-2 in the brain. Additional analysis revealed a significant decrease in the levels of COX2 and IL-1ß, but not in other proinflammatory cytokines. Importantly, long-term DHA supplementation significantly changed the n-3:n-6 polyunsaturated fatty acid ratio in the brain. CONCLUSIONS: Collectively, these data indicate that diet-induced accumulation of DHA in the brain protects against postischemic inflammation and injury. Because DHA is widely available at low cost and has an excellent safety profile, our data suggest that increased DHA intake may provide protection against acute immune response/brain damage in ischemic stroke.


Asunto(s)
Isquemia Encefálica/inmunología , Encéfalo/inmunología , Ácidos Docosahexaenoicos/administración & dosificación , Inmunidad Activa/inmunología , Neuronas/inmunología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Ácidos Docosahexaenoicos/metabolismo , Inflamación/inmunología , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Receptor Toll-Like 2/metabolismo
11.
J Neurochem ; 114(4): 1177-92, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20533992

RESUMEN

Peripherin is a type III intermediate filament protein that is up-regulated during neuronal injury and is a major component of pathological inclusions found within degenerating motor neurons of patients with amyotrophic lateral sclerosis (ALS). The relationship between these inclusions and their protein constituents remains largely unknown. We have previously shown that peripherin expression is characterized by tissue-specific, intra-isoform associations that contribute to filament structure; changes to the normal isoform expression pattern is associated with malformed filaments and intracellular inclusions. Here, we profile peripherin isoform expression and ratio changes in traumatic neuronal injury, transgenic mouse models of motor neuron disease, and ALS. Extensive western blot analyses of Triton X-100 soluble and insoluble fractions of neuronal tissue from these conditions revealed significant changes in peripherin isoform content which could be differentiated by electrophoretic banding patterns to produce distinct peripherin biochemical signatures. Significantly, we found that the pattern of peripherin expression in ALS most closely approximates that of peripherin over-expressing mice, but differs with regard to inter-individual variations in isoform-specific expression. Overall, these results provide important insights into complex post-transcriptional processes that may underlie a continuum between peripherin-mediated neuronal repair and its role in the pathogenesis of motor neuron disease.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas de Filamentos Intermediarios/metabolismo , Glicoproteínas de Membrana/metabolismo , Enfermedad de la Neurona Motora/genética , Enfermedad de la Neurona Motora/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Biomarcadores/metabolismo , Lesiones Encefálicas/etiología , Lesiones Encefálicas/fisiopatología , Regulación de la Expresión Génica/fisiología , Proteínas de Filamentos Intermediarios/biosíntesis , Proteínas de Filamentos Intermediarios/genética , Masculino , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Enfermedad de la Neurona Motora/fisiopatología , Compresión Nerviosa/métodos , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Periferinas , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Proteómica , Nervio Ciático/patología
12.
Stroke ; 39(3): 935-42, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18258827

RESUMEN

BACKGROUND AND PURPOSE: We sought to develop a model system for live analysis of brain inflammatory response in ischemic injury. METHODS: Using a reporter mouse-expressing luciferase gene under transcriptional control of the murine glial fibrillary acidic protein (GFAP) promoter (GFAP-luc mice) and biophotonic/bioluminescent imaging as tools, we developed a model system for in vivo analysis of astrocyte activation/response in cerebral ischemia. RESULTS: Analysis of photon emissions from the brains of living animals revealed marked sex differences in astrocyte response to ischemic injury. The increase in GFAP signals was significantly higher in female mice in the metestrus/diestrus period compared with male transgenic mice (1.71 x 10(7)+/-0.19 x 10(7) vs 0.92 x 10(7)+/-0.15 x 10(7), P<0.001). Similar results were obtained by quantitative immunohistochemistry (males vs females: 13.4+/-0.5 vs 16.96+/-0.64, P<0.0001). However, astrocyte activation/GFAP signals showed cyclic, estrus-dependent variations in response to ischemic injury. Physiologically higher levels of estrogen and application of pharmacologic doses of estrogen during replacement therapy attenuated GFAP upregulation after stroke. Interestingly, contrary to a positive correlation between the intensities of GFAP signals and infarct size in male mice, no such correlation was observed in any of the experimental groups of female GFAP-luc mice. CONCLUSIONS: Our results suggest that GFAP upregulation in ischemic injury may have different functional significance in female and male experimental animals and may not directly reflect the extent of ischemia-induced neuronal damage in female GFAP-luc mice. Using a novel live imaging approach, we demonstrated that the early-phase brain inflammatory response to ischemia may be associated with sex-specific biomarkers of brain damage.


Asunto(s)
Astrocitos/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Encefalitis/diagnóstico , Estrógenos/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Factores Sexuales , Animales , Isquemia Encefálica/complicaciones , Infarto Cerebral/etiología , Infarto Cerebral/patología , Diestro , Encefalitis/etiología , Encefalitis/metabolismo , Estrógenos/farmacología , Femenino , Proteína Ácida Fibrilar de la Glía/genética , Inmunohistoquímica , Luminiscencia , Masculino , Metestro , Ratones , Ratones Transgénicos , Regulación hacia Arriba/efectos de los fármacos
13.
J Neurosci ; 27(10): 2596-605, 2007 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-17344397

RESUMEN

Here we report in vivo evidence of a neuroprotective role of proliferating microglial cells in cerebral ischemia. Using transgenic mice expressing a mutant thymidine kinase form of herpes simplex virus driven by myeloid-specific CD11b promoter and ganciclovir treatment as a tool, we selectively ablated proliferating (Mac-2 positive) microglia after transient middle cerebral artery occlusion. The series of experiments using green fluorescent protein-chimeric mice demonstrated that within the first 72 h after ischemic injury, the Mac-2 marker [unlike Iba1 (ionized calcium-binding adapter molecule 1)] was preferentially expressed by the resident microglia. Selective ablation of proliferating resident microglia was associated with a marked alteration in the temporal dynamics of proinflammatory cytokine expression, a significant increase in the size of infarction associated with a 2.7-fold increase in the number of apoptotic cells, predominantly neurons, and a 1.8-fold decrease in the levels of IGF-1. A double-immunofluorescence analysis revealed a approximately 100% colocalization between IGF-1 positive cells and Mac-2, a marker of activated/proliferating resident microglia. Conversely, stimulation of microglial proliferation after cerebral ischemia by M-CSF (macrophage colony stimulating factor) resulted in a 1.9-fold increase in IGF-1 levels and a significant increase of Mac2+ cells. Our findings suggest that a postischemic proliferation of the resident microglial cells may serve as an important modulator of a brain inflammatory response. More importantly, our results revealed a marked neuroprotective potential of proliferating microglia serving as an endogenous pool of neurotrophic molecules such as IGF-1, which may open new therapeutic avenues in the treatment of stroke and other neurological disorders.


Asunto(s)
Isquemia Encefálica/patología , Encéfalo/patología , Proliferación Celular , Microglía/patología , Animales , Antivirales/farmacología , Apoptosis , Encéfalo/metabolismo , Antígeno CD11b/genética , Proliferación Celular/efectos de los fármacos , Infarto Cerebral/patología , Citocinas/metabolismo , Citoprotección , Galectina 3/metabolismo , Ganciclovir/farmacología , Mediadores de Inflamación/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor Estimulante de Colonias de Macrófagos/farmacología , Masculino , Ratones , Ratones Transgénicos/virología , Microglía/efectos de los fármacos , Microglía/metabolismo , Mutación , Neuronas/patología , Simplexvirus/genética , Timidina Quinasa/genética , Distribución Tisular
14.
Exp Neurol ; 204(1): 433-42, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17234187

RESUMEN

Considering that several pathways leading to cell death are activated in cerebral ischemia, we tested in mouse models of transient and permanent ischemia a drug cocktail aiming at distinct pharmacological targets during the evolution of ischemic injury. It consists of minocycline--an antibiotic with anti-inflammatory properties, riluzole--a glutamate antagonist, and nimodipine--a blocker of voltage-gated calcium channels. Administered 2 h after transient or permanent MCAO, it significantly decreased the size of infarction, by approximately 65% after transient and approximately 35% after permanent ischemia and markedly improve clinical recovery of mice. In both experimental models a three-drug cocktail achieved significantly more efficient neuroprotection than any of the components tested alone. However, some interesting observation emerged from the single-drug studies. Treatment with minocycline alone was efficient in both experimental models while treatment with glutamate antagonist riluzole conferred neuroprotection only after transient MCAO. Immunohistochemical analysis following three-drug treatment revealed reduced microglia/macrophages and caspase-3 activation as well as preserved GFAP immunoreactivity following transient ischemia. No detectable differences in the levels of Mac-2, GFAP and caspase-3 immunoreactivities were observed 72 h after permanent MCAO. These marked differences in the brain tissue responses to ischemic injury and to treatments suggest that different pathological mechanisms may be operating in transient and permanent ischemia. However, the three-drug cocktail exerted significant neuroprotection in both experimental models thus demonstrating that simultaneous targeting of several pathophysiological pathways involved in the evolution of ischemic injury may represent a rational therapeutic strategy for stroke.


Asunto(s)
Isquemia Encefálica/patología , Ataque Isquémico Transitorio/patología , Minociclina/farmacología , Fármacos Neuroprotectores/farmacología , Nimodipina/farmacología , Riluzol/farmacología , Animales , Encéfalo/enzimología , Encéfalo/patología , Caspasa 3/metabolismo , Combinación de Medicamentos , Activación Enzimática , Proteína Ácida Fibrilar de la Glía/metabolismo , Inmunohistoquímica , Ataque Isquémico Transitorio/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/patología , Infarto del Miocardio/patología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...