Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Neurobiol Dis ; 134: 104683, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31765727

RESUMEN

Repeated mild traumatic brain injury (rmTBI) can lead to development of chronic traumatic encephalopathy (CTE), which is characterized by progressive neurodegeneration with presence of white matter damage, gliosis and hyper-phosphorylated tau. While animal models of rmTBI have been documented, few characterize the molecular pathogenesis and expression profiles of relevant injured brain regions. Additionally, while the usage of transgenic tau mice in rmTBI is prevalent, the effects of tau on pathological outcomes has not been well studied. Here we characterized a 42-impact closed-head rmTBI paradigm on 3-4 month old male C57BL/6 (WT) and Tau-overexpressing mice (Tau58.4). This injury paradigm resulted in chronic gliosis, T-cell infiltration, and demyelination of the optic nerve and associated white matter tracts at 1-month post-injury. At 3-months post-injury, Tau58.4 mice showed progressive neuroinflammation and neurodegeneration in multiple brain regions compared to WT mice. Corresponding to histopathology, RNAseq of the optic nerve tract at 1-month post-injury showed significant upregulation of inflammatory pathways and downregulation of myelin synthetic pathways in both genotypes. However, Tau58.4 mice showed additional changes in neurite development, protein processing, and cell stress. Comparisons with published transcriptomes of human Alzheimer's Disease and CTE revealed common signatures including neuroinflammation and downregulation of protein phosphatases. We next investigated the demyelination and T-cell infiltration phenotypes to determine whether these offer potential avenues for therapeutic intervention. Tau58.4 mice were treated with the histamine H3 receptor antagonist GSK239512 for 1-month post-injury to promote remyelination of white matter lesions. This restored myelin gene expression to sham levels but failed to repair the histopathologic lesions. Likewise, injured T-cell-deficient Rag2/Il2rg (R2G2) mice also showed evidence for inflammation and loss of myelin. However, unlike immune-competent mice, R2G2 mice had altered myeloid cell gene expression and fewer demyelinated lesions. Together this data shows that rmTBI leads to chronic white matter inflammatory demyelination and axonal loss exacerbated by human tau overexpression but suggests that immune-suppression and remyelination alone are insufficient to reverse damage.


Asunto(s)
Conmoción Encefálica/metabolismo , Conmoción Encefálica/patología , Encéfalo/metabolismo , Encéfalo/patología , Proteínas tau/metabolismo , Animales , Conmoción Encefálica/complicaciones , Encefalitis/complicaciones , Encefalitis/metabolismo , Encefalitis/patología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Sustancia Blanca/metabolismo , Sustancia Blanca/patología
2.
PLoS One ; 13(11): e0206934, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30395590

RESUMEN

Epidemiological studies have established an association between air pollution particulate matter exposure (PM2.5) and neurocognitive decline. Experimental data suggest that microglia play an essential role in air pollution PM-induced neuroinflammation and oxidative stress. This study examined the effect of nano-sized particulate matter (nPM) on complement C5 deposition and microglial activation in the corpus callosum of mice (C57BL/6J males). nPM was collected in an urban Los Angeles region impacted by traffic emissions. Mice were exposed to 10 weeks of re-aerosolized nPM or filtered air for a cumulative 150 hours. nPM-exposed mice exhibited reactive microglia and 2-fold increased local deposition of complement C5/ C5α proteins and complement component C5a receptor 1 (CD88) in the corpus callosum. However, serum C5 levels did not differ between nPM and filtered air cohorts. These findings demonstrate white matter C5 deposition and microglial activation secondary to nPM exposure. The C5 upregulation appears to be localized to the brain.


Asunto(s)
Contaminación del Aire/efectos adversos , Complemento C5/genética , Inflamación/fisiopatología , Trastornos Neurocognitivos/fisiopatología , Animales , Cuerpo Calloso/efectos de los fármacos , Cuerpo Calloso/patología , Humanos , Inflamación/inducido químicamente , Inflamación/genética , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Nanopartículas/efectos adversos , Trastornos Neurocognitivos/inducido químicamente , Trastornos Neurocognitivos/genética , Estrés Oxidativo/efectos de los fármacos , Material Particulado/efectos adversos
3.
Neurol Res ; 39(10): 910-917, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28828966

RESUMEN

OBJECTIVES: Chronic cerebral hypoperfusion (CCH) can result in vascular dementia and small vessel white matter ischemic injury. These findings have previously been demonstrated in a murine experimental model of CCH secondary to bilateral common carotid artery stenosis (BCAS). This study sought to elucidate the effects of CCH on recognition memory as assessed by the novel object recognition (NOR) test and histological analysis of the hippocampus and perirhinal cortex. METHODS: Studies were performed on ten-week-old male mice using bilateral 0.18 mm microcoils to narrow the carotid arteries in accordance with prior publications. Following surgery, BCAS (n = 6) and sham (n = 6) mice were evaluated using NOR and 8-arm radial maze testing paradigms. Tissue damage was assessed using H&E staining on a parallel cohort of mice (n = 6 BCAS, n = 7 sham). RESULTS: In the NOR paradigm, BCAS mice demonstrated significant deficits in short-term memory. Consistent with prior studies, BCAS mice also performed significantly worse on 8-arm radial maze testing. BCAS mice exhibited significantly more neuronal injury in the perirhinal cortex when compared to sham-operated mice. However, no significant differences in neuronal damage were observed in the CA1 region of the hippocampus. DISCUSSION: Experimental CCH secondary to BCAS results in recognition memory deficits on NOR testing. Damage to the perirhinal cortex, rather than to the hippocampus, may underlie this impairment.


Asunto(s)
Estenosis Carotídea/fisiopatología , Estenosis Carotídea/psicología , Demencia Vascular/fisiopatología , Reconocimiento en Psicología/fisiología , Animales , Región CA1 Hipocampal/irrigación sanguínea , Región CA1 Hipocampal/patología , Región CA1 Hipocampal/fisiopatología , Estenosis Carotídea/complicaciones , Estenosis Carotídea/patología , Circulación Cerebrovascular/fisiología , Enfermedad Crónica , Demencia Vascular/etiología , Demencia Vascular/patología , Modelos Animales de Enfermedad , Masculino , Aprendizaje por Laberinto/fisiología , Memoria a Corto Plazo/fisiología , Ratones Endogámicos C57BL , Neuronas/patología , Neuronas/fisiología , Corteza Perirrinal/irrigación sanguínea , Corteza Perirrinal/patología , Corteza Perirrinal/fisiopatología
4.
Environ Health Perspect ; 124(10): 1537-1546, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27187980

RESUMEN

BACKGROUND: Rodent models for urban air pollution show consistent induction of inflammatory responses in major brain regions. However, the initial impact of air pollution particulate material on olfactory gateways has not been reported. OBJECTIVE: We evaluated the olfactory neuroepithelium (OE) and brain regional responses to a nanosized subfraction of urban traffic ultrafine particulate matter (nPM, < 200 nm) in vivo, ex vivo, and in vitro. METHODS: Adult mice were exposed to reaerosolized nPM for 5, 20, and 45 cumulative hours over 3 weeks. The OE, the olfactory bulb (OB), the cerebral cortex, and the cerebellum were analyzed for oxidative stress and inflammatory responses. Acute responses of the OE to liquid nPM suspensions were studied with ex vivo and primary OE cultures. RESULTS: After exposure to nPM, the OE and OB had rapid increases of 4-hydroxy-2-nonenal (4-HNE) and 3-nitrotyrosine (3-NT) protein adducts, whereas the cerebral cortex and cerebellum did not respond at any time. All brain regions showed increased levels of tumor necrosis factor-α (TNFα) protein by 45 hr, with earlier induction of TNFα mRNA in OE and OB. These responses corresponded to in vitro OE and mixed glial responses, with rapid induction of nitrite and inducible nitric oxide synthase (iNOS), followed by induction of TNFα. CONCLUSIONS: These findings show the differential time course of oxidative stress and inflammatory responses to nPM between the OE and the brain. Slow cumulative transport of inhaled nPM into the brain may contribute to delayed responses of proximal and distal brain regions, with potential input from systemic factors. CITATION: Cheng H, Saffari A, Sioutas C, Forman HJ, Morgan TE, Finch CE. 2016. Nanoscale particulate matter from urban traffic rapidly induces oxidative stress and inflammation in olfactory epithelium with concomitant effects on brain. Environ Health Perspect 124:1537-1546; http://dx.doi.org/10.1289/EHP134.

5.
PLoS One ; 11(4): e0153376, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27071057

RESUMEN

This study examines the effects of nano-size particulate matter (nPM) exposure in the setting of murine reperfused stroke. Particulate matter is a potent source of inflammation and oxidative stress. These processes are known to influence stroke progression through recruitment of marginally viable penumbral tissue into the ischemic core. nPM was collected in an urban area in central Los Angeles, impacted primarily by traffic emissions. Re-aerosolized nPM or filtered air was then administered to mice through whole body exposure chambers for forty-five cumulative hours. Exposed mice then underwent middle cerebral artery occlusion/ reperfusion. Following cerebral ischemia/ reperfusion, mice exposed to nPM exhibited significantly larger infarct volumes and less favorable neurological deficit scores when compared to mice exposed to filtered air. Mice exposed to nPM also demonstrated increases in markers of inflammation and oxidative stress in the region of the ischemic core. The findings suggest a detrimental effect of urban airborne particulate matter exposure in the setting of acute ischemic stroke.


Asunto(s)
Isquemia Encefálica/fisiopatología , Nanopartículas/efectos adversos , Material Particulado/efectos adversos , Accidente Cerebrovascular/fisiopatología , Animales , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/fisiopatología , Inflamación/inducido químicamente , Los Angeles , Ratones , Estrés Oxidativo/efectos de los fármacos , Tamaño de la Partícula , Reperfusión/métodos
6.
J Neuroinflammation ; 13: 19, 2016 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-26810976

RESUMEN

BACKGROUND: The basis for air pollution-associated neurodegenerative changes in humans is being studied in rodent models. We and others find that the ultrafine particulate matter (PM) derived from vehicular exhaust can induce synaptic dysfunction and inflammatory responses in vivo and in vitro. In particular, a nano-sized subfraction of particulate matter (nPM, PM0.2) from a local urban traffic corridor can induce glial TNFα production in mixed glia (astrocytes and microglia) derived from neonatal rat cerebral cortex. METHODS: Here, we examine the role of TNFα in neurite dysfunctions induced by nPM in aqueous suspensions at 12 µg/ml. First, we show that the proximal brain gateway to nPM, the olfactory neuroepithelium (OE), rapidly responds to nPM ex vivo, with induction of TNFα, activation of macrophages, and dendritic shrinkage. Cell interactions were further analyzed with mixed glia and neurons from neonatal rat cerebral cortex. RESULTS: Microglia contributed more than astrocytes to TNFα induction by nPM. We then showed that the threefold higher TNFα in conditioned media (nPM-CM) from mixed glia was responsible for the inhibition of neurite outgrowth by small interfering RNA (siRNA) TNFα knockdown and by TNFα immunoneutralization. Despite lack of TNFR1 induction by nPM in the OE, experimental blocking of TNFR1 by TNFα receptor blockers restored total neurite length. CONCLUSIONS: These findings implicate microglia-derived TNFα as a mediator of nPM in air pollution-associated neurodegenerative changes which alter synaptic functions and neuronal growth.


Asunto(s)
Neuritas/efectos de los fármacos , Neuroglía/citología , Material Particulado/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Emisiones de Vehículos , Animales , Animales Recién Nacidos , Células Cultivadas , Corteza Cerebral/citología , Citocinas/genética , Citocinas/metabolismo , Epitelio/metabolismo , Femenino , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuroglía/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Bulbo Olfatorio/citología , Bulbo Olfatorio/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Tubulina (Proteína)/metabolismo , Factor de Necrosis Tumoral alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...