Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Neuroinflammation ; 18(1): 256, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34740378

RESUMEN

BACKGROUND: Neurotoxic microglia and astrocytes begin to activate and participate in pathological processes after spinal cord injury (SCI), subsequently causing severe secondary damage and affecting tissue repair. We have previously reported that photobiomodulation (PBM) can promote functional recovery by reducing neuroinflammation after SCI, but little is known about the underlying mechanism. Therefore, we aimed to investigate whether PBM ameliorates neuroinflammation by modulating the activation of microglia and astrocytes after SCI. METHODS: Male Sprague-Dawley rats were randomly divided into three groups: a sham control group, an SCI + vehicle group and an SCI + PBM group. PBM was performed for two consecutive weeks after clip-compression SCI models were established. The activation of neurotoxic microglia and astrocytes, the level of tissue apoptosis, the number of motor neurons and the recovery of motor function were evaluated at different days post-injury (1, 3, 7, 14, and 28 days post-injury, dpi). Lipocalin 2 (Lcn2) and Janus kinase-2 (JAK2)-signal transducer and activator of transcription-3 (STAT3) signaling were regarded as potential targets by which PBM affected neurotoxic microglia and astrocytes. In in vitro experiments, primary microglia and astrocytes were irradiated with PBM and cotreated with cucurbitacin I (a JAK2-STAT3 pathway inhibitor), an adenovirus (shRNA-Lcn2) and recombinant Lcn2 protein. RESULTS: PBM promoted the recovery of motor function, inhibited the activation of neurotoxic microglia and astrocytes, alleviated neuroinflammation and tissue apoptosis, and increased the number of neurons retained after SCI. The upregulation of Lcn2 and the activation of the JAK2-STAT3 pathway after SCI were suppressed by PBM. In vitro experiments also showed that Lcn2 and JAK2-STAT3 were mutually promoted and that PBM interfered with this interaction, inhibiting the activation of microglia and astrocytes. CONCLUSION: Lcn2/JAK2-STAT3 crosstalk is involved in the activation of neurotoxic microglia and astrocytes after SCI, and this process can be suppressed by PBM.


Asunto(s)
Astrocitos/efectos de la radiación , Terapia por Luz de Baja Intensidad , Microglía/efectos de la radiación , Recuperación de la Función/efectos de la radiación , Traumatismos de la Médula Espinal/patología , Animales , Astrocitos/metabolismo , Janus Quinasa 2/metabolismo , Janus Quinasa 2/efectos de la radiación , Lipocalina 2/metabolismo , Lipocalina 2/efectos de la radiación , Masculino , Microglía/metabolismo , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/patología , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/efectos de la radiación , Transducción de Señal/efectos de la radiación , Traumatismos de la Médula Espinal/metabolismo , Regulación hacia Arriba
2.
Theranostics ; 11(3): 1269-1294, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33391534

RESUMEN

Neonatal hypoxic-ischemic (HI) injury is a severe complication often leading to neonatal death and long-term neurobehavioral deficits in children. Currently, the only treatment option available for neonatal HI injury is therapeutic hypothermia. However, the necessary specialized equipment, possible adverse side effects, and limited effectiveness of this therapy creates an urgent need for the development of new HI treatment methods. Photobiomodulation (PBM) has been shown to be neuroprotective against multiple brain disorders in animal models, as well as limited human studies. However, the effects of PBM treatment on neonatal HI injury remain unclear. Methods: Two-minutes PBM (808 nm continuous wave laser, 8 mW/cm2 on neonatal brain) was applied three times weekly on the abdomen of pregnant rats from gestation day 1 (GD1) to GD21. After neonatal right common carotid artery ligation, cortex- and hippocampus-related behavioral deficits due to HI insult were measured using a battery of behavioral tests. The effects of HI insult and PBM pretreatment on infarct size; synaptic, dendritic, and white matter damage; neuronal degeneration; apoptosis; mitochondrial function; mitochondrial fragmentation; oxidative stress; and gliosis were then assessed. Results: Prenatal PBM treatment significantly improved the survival rate of neonatal rats and decreased infarct size after HI insult. Behavioral tests revealed that prenatal PBM treatment significantly alleviated cortex-related motor deficits and hippocampus-related memory and learning dysfunction. In addition, mitochondrial function and integrity were protected in HI animals treated with PBM. Additional studies revealed that prenatal PBM treatment significantly alleviated HI-induced neuroinflammation, oxidative stress, and myeloid cell/astrocyte activation. Conclusion: Prenatal PBM treatment exerts neuroprotective effects on neonatal HI rats. Underlying mechanisms for this neuroprotection may include preservation of mitochondrial function, reduction of inflammation, and decreased oxidative stress. Our findings support the possible use of PBM treatment in high-risk pregnancies to alleviate or prevent HI-induced brain injury in the perinatal period.


Asunto(s)
Hipoxia-Isquemia Encefálica/radioterapia , Hipoxia/radioterapia , Isquemia/radioterapia , Animales , Animales Recién Nacidos , Apoptosis/efectos de la radiación , Astrocitos/efectos de la radiación , Corteza Cerebral/efectos de la radiación , Modelos Animales de Enfermedad , Femenino , Hipocampo/efectos de la radiación , Terapia por Luz de Baja Intensidad/métodos , Masculino , Mitocondrias/efectos de la radiación , Neuronas/efectos de la radiación , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo/efectos de la radiación , Embarazo , Ratas , Ratas Sprague-Dawley
3.
Cell Mol Neurobiol ; 40(1): 141-152, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31446561

RESUMEN

Spinal cord injury (SCI) stimulates reactive astrogliosis and the infiltration of macrophages, which interact with each other at the injured area. We previously found Photobiomodulation (PBM) significantly decreases the number of M1 macrophages at the injured area of SCI. But the exact nature of the astrocyte response following PBM and relationship with the macrophage have not been explored in detail. In this study, a BALB/c mice model with standardized bilateral spinal cord compression and a macrophage-astrocyte co-culture model were applied to study effects of PBM on astrocytes. Results showed that PBM inhibit the expression of the astrocyte markers glial fibrillary acidic protein (GFAP) and the secretion of chondroitin sulfate proteoglycans (CSPG) in the para-epicenter area, decrease the number of M1 macrophage in vivo. The in vitro experiments indicated M1 macrophages promote the cell viability of astrocytes and the expression of CSPG. However, PBM significantly inhibited the expression of GFAP, decreased activation of astrocyte, and downregulated the expression of CSPG by regulating M1 macrophages. These results demonstrate that PBM may regulate the interaction between macrophages and astrocytes after spinal cord injury, which inhibited the formation of glial scar.


Asunto(s)
Astrocitos/efectos de la radiación , Polaridad Celular/efectos de la radiación , Terapia por Luz de Baja Intensidad , Macrófagos/efectos de la radiación , Animales , Astrocitos/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Medios de Cultivo Condicionados/farmacología , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Macrófagos/efectos de los fármacos , Ratones Endogámicos BALB C , Actividad Motora/efectos de los fármacos , Actividad Motora/efectos de la radiación , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , Recuperación de la Función/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Traumatismos de la Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/radioterapia
4.
Sci Rep ; 9(1): 9588, 2019 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-31270437

RESUMEN

Better survival rates among pediatric brain tumor patients have resulted in an increased awareness of late side effects that commonly appear following cancer treatment. Radiation-induced changes in hippocampus and white matter are well described, but do not explain the full range of neurological late effects in childhood cancer survivors. The aim of this study was to investigate thalamus following cranial irradiation (CIR) to the developing brain. At postnatal day 14, male mice pups received a single dose of 8 Gy CIR. Cellular effects in thalamus were assessed using immunohistochemistry 4 months after CIR. Interestingly, the density of neurons decreased with 35% (p = 0.0431) and the density of astrocytes increased with 44% (p = 0.011). To investigate thalamic astrocytes, S100ß+ cells were isolated by fluorescence-activated cell sorting and genetically profiled using next-generation sequencing. The phenotypical characterization indicated a disrupted function, such as downregulated microtubules' function, higher metabolic activity, immature phenotype and degraded ECM. The current study provides novel insight into that thalamus, just like hippocampus and white matter, is severely affected by CIR. This knowledge is of importance to understand the late effects seen in pediatric brain tumor survivors and can be used to give them the best suitable care.


Asunto(s)
Irradiación Craneana , Radiación Ionizante , Tálamo/efectos de la radiación , Animales , Astrocitos/citología , Astrocitos/metabolismo , Astrocitos/efectos de la radiación , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de la radiación , Secuenciación de Nucleótidos de Alto Rendimiento , Masculino , Ratones , Ratones Endogámicos C57BL , Microtúbulos/metabolismo , Fenotipo , Análisis de Secuencia de ADN , Tálamo/metabolismo , Tálamo/patología
5.
J Biophotonics ; 10(12): 1761-1771, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28464523

RESUMEN

We aimed to investigate the effects of low-level light emitting diode therapy (LED-T) on the long-term functional outcomes after cerebral ischemia, and the optimal timing of LED-T initiation for achieving suitable functional recovery. Focal cerebral ischemia was induced in mice via photothrombosis. These mice were assigned to a sham-operated (control), ischemic (vehicle), or LED-T group [initiation immediately (acute), 4 days (subacute) or 10 days (delayed) after ischemia, followed by once-daily treatment for 7 days]. Behavioral outcomes were assessed 21 and 28 days post-ischemia, and histopathological analysis was performed 28 days post-ischemia. The acute and subacute LED-T groups showed a significant improvement in motor function up to 28 days post-ischemia, although no brain atrophy recovery was noted. We observed proliferating cells (BrdU+ ) in the ischemic brain, and significant increases in BrdU+ /GFAP+ , BrdU+ /DCX+ , BrdU+ /NeuN+ , and CD31+ cells in the subacute LED-T group. However, the BrdU+ /Iba-1+ cell count was reduced in the subacute LED-T group. Furthermore, the brain-derived neurotrophic factor (BDNF) was significantly upregulated in the subacute LED-T group. We concluded that LED-T administered during the subacute stage had a positive impact on the long-term functional outcome, probably via neuron and astrocyte proliferation, blood vessel reconstruction, and increased BDNF expression. Picture: The rotarod test for motor coordination showed that acute and subacute LED-T improves long-term functional recovery after cerebral ischemia.


Asunto(s)
Isquemia Encefálica/fisiopatología , Isquemia Encefálica/terapia , Fototerapia/instrumentación , Recuperación de la Función/efectos de la radiación , Accidente Cerebrovascular/fisiopatología , Accidente Cerebrovascular/terapia , Animales , Astrocitos/patología , Astrocitos/efectos de la radiación , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Diferenciación Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Modelos Animales de Enfermedad , Proteína Doblecortina , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/patología , Microglía/efectos de la radiación , Neuronas/patología , Neuronas/efectos de la radiación , Factores de Tiempo
6.
Bioelectromagnetics ; 37(5): 338-50, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27272062

RESUMEN

The widespread use of mobile phones by adolescents raises concerns about possible health effects of radiofrequency electromagnetic fields (RF EMF 900 MHz) on the immature brain. Neuro-development is a period of particular sensitivity to repeated environmental challenges such as pro-inflammatory insults. Here, we used rats to assess whether astrocyte reactivity, perception, and emotionality were affected by RF EMF exposures during adolescence. We also investigated if adolescent brains were more sensitive to RF EMF exposures after neurodevelopmental inflammation. To do so, we either performed 80 µg/kg intra-peritoneal injections of lipopolysaccharides during gestation or 1.25 µg/h intra-cerebro-ventricular infusions during adolescence. From postnatal day (P)32 to 62, rats were subjected to 45 min RF EMF exposures to the brain (specific absorption rates: 0, 1.5, or 6 W/kg, 5 days/week). From P56, they were tested for perception of novelty, anxiety-like behaviors, and emotional memory. To assess astrocytic reactivity, Glial Fibrillary Acidic Protein was measured at P64. Our results did not show any neurobiological impairment in healthy and vulnerable RF EMF-exposed rats compared to their sham-exposed controls. These data did not support the hypothesis of a specific cerebral sensitivity to RF EMF of adolescents, even after a neurodevelopmental inflammation. Bioelectromagnetics. 37:338-350, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Astrocitos/efectos de la radiación , Encéfalo/fisiopatología , Encéfalo/efectos de la radiación , Efectos Tardíos de la Exposición Prenatal/patología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Ondas de Radio/efectos adversos , Adolescente , Animales , Ansiedad/etiología , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/patología , Conducta Animal/efectos de los fármacos , Conducta Animal/efectos de la radiación , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Modelos Animales de Enfermedad , Emociones/efectos de los fármacos , Emociones/efectos de la radiación , Femenino , Humanos , Lipopolisacáridos/farmacología , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/psicología , Ratas
7.
Int J Neurosci ; 126(1): 76-87, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-25469453

RESUMEN

We have used the MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) mouse model to explore whether (i) the neuroprotective effect of near infrared light (NIr) treatment in the SNc is dose-dependent and (ii) the relationship between tyrosine hydroxylase (TH)+ terminal density and glial cells in the caudate-putamen complex (CPu). Mice received MPTP injections (50 mg/kg) and 2 J/cm2 NIr dose with either 2 d or 7 d survival period. In another series, with a longer 14 d survival period, mice had a stronger MPTP regime (100 mg/kg) and either 2 J/cm2 or 4 J/cm2 NIr dose. Brains were processed for routine immunohistochemistry and cell counts were made using stereology. Our findings were that in the 2 d series, no change in SNc TH+ cell number was evident after any treatment. In the 7 d series however, MPTP insult resulted in ∼45% reduction in TH+ cell number; after NIr (2 J/cm2) treatment, many cells were protected from the toxic insult. In the 14 d series, MPTP induced a similar reduction in TH+ cell number. NIr mitigated the loss of TH+ cells, but only at the higher dose of 4 J/cm2; the lower dose of 2 J/cm2 had no neuroprotective effect in this series. The higher dose of NIr, unlike the lower dose, also mitigated the MPTP- induced increase in CPu astrocytes after 14 d; these changes were independent of TH+ terminal density, of which, did not vary across the different experimental groups. In summary, we showed that neuroprotection by NIr irradiation in MPTP-treated mice was dose-dependent; with increasing MPTP toxicity, higher doses of NIr were required to protect cells and reduce astrogliosis.


Asunto(s)
Neuronas Dopaminérgicas/efectos de la radiación , Gliosis/radioterapia , Rayos Infrarrojos/uso terapéutico , Intoxicación por MPTP/radioterapia , Trastornos Parkinsonianos/radioterapia , Porción Compacta de la Sustancia Negra/efectos de la radiación , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/administración & dosificación , Animales , Astrocitos/patología , Astrocitos/efectos de la radiación , Núcleo Caudado/patología , Núcleo Caudado/efectos de la radiación , Recuento de Células , Supervivencia Celular/efectos de la radiación , Neuronas Dopaminérgicas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Gliosis/patología , Terapia por Luz de Baja Intensidad , Intoxicación por MPTP/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas del Tejido Nervioso/análisis , Trastornos Parkinsonianos/patología , Porción Compacta de la Sustancia Negra/patología , Putamen/patología , Putamen/efectos de la radiación , Tirosina 3-Monooxigenasa/análisis
8.
Nat Commun ; 5: 3262, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24500276

RESUMEN

Astrocytes play important roles in synaptic transmission and plasticity. Despite in vitro evidence, their causal contribution to cortical network activity and sensory information processing in vivo remains unresolved. Here we report that selective photostimulation of astrocytes with channelrhodopsin-2 in primary visual cortex enhances both excitatory and inhibitory synaptic transmission, through the activation of type 1a metabotropic glutamate receptors. Photostimulation of astrocytes in vivo increases the spontaneous firing of parvalbumin-positive (PV(+)) inhibitory neurons, while excitatory and somatostatin-positive (SOM(+)) neurons show either an increase or decrease in their activity. Moreover, PV(+) neurons show increased baseline visual responses and reduced orientation selectivity to visual stimuli, whereas excitatory and SOM(+) neurons show either increased or decreased baseline visual responses together with complementary changes in orientation selectivity. Therefore, astrocyte activation, through the dual control of excitatory and inhibitory drive, influences neuronal integrative features critical for sensory information processing.


Asunto(s)
Astrocitos/efectos de la radiación , Receptores de Glutamato Metabotrópico/metabolismo , Corteza Visual/fisiología , Animales , Astrocitos/metabolismo , Calcio/metabolismo , Células Cultivadas , Channelrhodopsins , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Optogenética , Transmisión Sináptica
9.
Nanomedicine ; 10(1): 15-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24200521

RESUMEN

Alzheimer's disease (AD) is the sixth leading cause of age-related death with no effective intervention yet available. Our previous studies have demonstrated the potential efficacy of Low Level Laser Therapy (LLLT) in AD cell models by mitigating amyloid-ß peptide (Aß)-induced oxidative stress and inflammation. However, the penetration depth of light is still the major challenge for implementing LLLT in animal models and in the clinical settings. In this study, we present the potential of applying Bioluminescence Resonance Energy Transfer to Quantum Dots (BRET-Qdots) as an alternative near infrared (NIR) light source for LLLT. Our results show that BRET-Qdot-emitted NIR suppresses Aß-induced oxidative stress and inflammatory responses in primary rat astrocytes. These data provide a proof of concept for a nanomedicine platform for LLLT. FROM THE CLINICAL EDITOR: Low Level Laser Therapy has already been demonstrated to mitigate amyloid-ß peptide induced oxidative stress and inflammation, a key driver of Alzheimer's disease. The major issue in moving this forward from cell cultures to live animals and potentially to human subjects is light penetration depth. In this novel study, BRET-Qdots were used as an alternative near infrared light source with good efficacy, paving the way to the development of a nanomedicine platform.


Asunto(s)
Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/metabolismo , Terapia por Luz de Baja Intensidad , Nanopartículas/química , Estrés Oxidativo , Enfermedad de Alzheimer/patología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Astrocitos/efectos de la radiación , Transferencia de Energía por Resonancia de Bioluminiscencia , Humanos , Inflamación/patología , Inflamación/terapia , Luz , Nanomedicina , Nanopartículas/administración & dosificación , Puntos Cuánticos/uso terapéutico , Ratas
10.
Free Radic Biol Med ; 65: 1060-1068, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24012919

RESUMEN

About 500,000 new cancer patients will develop brain metastases in 2013. The primary treatment modality for these patients is partial or whole brain irradiation which leads to a progressive, irreversible cognitive impairment. Although the exact mechanisms behind this radiation-induced brain injury are unknown, neuroinflammation in glial populations is hypothesized to play a role. Blockers of the renin-angiotensin system (RAS) prevent radiation-induced cognitive impairment and modulate radiation-induced neuroinflammation. Recent studies suggest that RAS blockers may reduce inflammation by increasing endogenous concentrations of the anti-inflammatory heptapeptide angiotensin-(1-7) [Ang-(1-7)]. Ang-(1-7) binds to the AT(1-7) receptor and inhibits MAP kinase activity to prevent inflammation. This study describes the inflammatory response to radiation in astrocytes characterized by radiation-induced increases in (i) IL-1ß and IL-6 gene expression; (ii) COX-2 and GFAP immunoreactivity; (iii) activation of AP-1 and NF-κB transcription factors; and (iv) PKCα, MEK, and ERK (MAP kinase) activation. Treatment with U-0126, a MEK inhibitor, demonstrates that this radiation-induced inflammation in astrocytes is mediated through the MAP kinase pathway. Ang-(1-7) inhibits radiation-induced inflammation, increases in PKCα, and MAP kinase pathway activation (phosphorylation of MEK and ERK). Additionally Ang-(1-7) treatment leads to an increase in dual specificity phosphatase 1 (DUSP1). Furthermore, treatment with sodium vanadate (Na3VO4), a phosphatase inhibitor, blocks Ang-(1-7) inhibition of radiation-induced inflammation and MAP kinase activation, suggesting that Ang-(1-7) alters phosphatase activity to inhibit radiation-induced inflammation. These data suggest that RAS blockers inhibit radiation-induced inflammation and prevent radiation-induced cognitive impairment not only by reducing Ang II but also by increasing Ang-(1-7) levels.


Asunto(s)
Angiotensina I/farmacología , Astrocitos/inmunología , Sistema de Señalización de MAP Quinasas , Fragmentos de Péptidos/farmacología , Protectores contra Radiación/farmacología , Animales , Astrocitos/efectos de la radiación , Células Cultivadas , Evaluación Preclínica de Medicamentos , Fosfatasa 1 de Especificidad Dual/metabolismo , Inflamación/metabolismo , Cultivo Primario de Células , Ratas
11.
Neuroscience ; 171(3): 859-68, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20884337

RESUMEN

Oxidative stress and inflammation are important processes in the progression of Alzheimer's disease (AD). Recent studies have implicated the role of amyloid ß-peptides (Aß) in mediating these processes. In astrocytes, oligomeric Aß induces the assembly of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complexes resulting in its activation to produce anionic superoxide. Aß also promotes production of pro-inflammatory factors in astrocytes. Since low energy laser has previously been reported to attenuate oxidative stress and inflammation in biological systems, the objective of this study was to examine whether this type of laser light was able to abrogate the oxidative and inflammatory responses induced by Aß. Primary rat astrocytes were exposed to Helium-Neon laser (λ=632.8 nm), followed by the treatment with oligomeric Aß. Primary rat astrocytes were used to measure Aß-induced production of superoxide anions using fluorescence microscopy of dihydroethidium (DHE), assembly of NADPH oxidase subunits by the colocalization between the cytosolic p47(phox) subunit and the membrane gp91(phox) subunit using fluorescent confocal microscopy, phosphorylation of cytosolic phospholipase A(2) cPLA(2) and expressions of pro-inflammatory factors including interleukin-1ß (IL-1ß) and inducible nitric-oxide synthase (iNOS) using Western blot Analysis. Our data showed that laser light at 632.8 nm suppressed Aß-induced superoxide production, colocalization between NADPH oxidase gp91(phox) and p47(phox) subunits, phosphorylation of cPLA(2,) and the expressions of IL-1ß and iNOS in primary astrocytes. We demonstrated for the first time that 632.8 nm laser was capable of suppressing cellular pathways of oxidative stress and inflammatory responses critical in the pathogenesis in AD. This study should prove to provide the groundwork for further investigations for the potential use of laser therapy as a treatment for AD.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/efectos de la radiación , Astrocitos/patología , Astrocitos/efectos de la radiación , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/toxicidad , Terapia por Luz de Baja Intensidad/métodos , Estrés Oxidativo/efectos de la radiación , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/efectos de la radiación , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/toxicidad , Animales , Animales Recién Nacidos , Células Cultivadas , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Corteza Cerebral/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Mediadores de Inflamación/efectos de la radiación , Estrés Oxidativo/fisiología , Fragmentos de Péptidos/toxicidad , Ratas , Superóxidos/antagonistas & inhibidores , Superóxidos/metabolismo
12.
Strahlenther Onkol ; 183(5): 248-55, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17497096

RESUMEN

BACKGROUND: Little is known about the immediate effects of whole-brain gamma-irradiation. The authors hypothesize that Egr1 as an immediate early gene and microglia both participate in early reactions. MATERIAL AND METHODS: Both, expression of Egr1 and cellular distribution were studied in a temporal sequence in different brain regions of rats subjected to irradiation with 10 Gy. Brain tissue was examined using immunohistochemistry, real-time RT-PCR (reverse transcription-polymerase chain reaction), and Western blotting. RESULTS: Astroglia and oligodendroglia showed increased Egr1 immunoreactivity within the first hours following irradiation. This was accompanied by a strong peak in CD68 immunoreactivity histologically attributable to activated microglia. A high constitutive expression of Egr1 protein in the nuclei of activated neurons was reduced following irradiation and RT-PCR demonstrated significantly reduced levels of egr1-lv as a neuronal activity-related mRNA variant. CONCLUSION: The induction of Egr1 in glial cells, as well as the activation of microglia take place earlier than histological changes reported so far. The authors revealed a temporal sequence of reactions that point toward the initiation of an immediate inflammatory response including reduced neuronal activity.


Asunto(s)
Encéfalo/efectos de la radiación , Irradiación Craneana/efectos adversos , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Microglía/efectos de la radiación , ARN Mensajero/genética , Traumatismos Experimentales por Radiación/genética , Animales , Antígenos CD/genética , Antígenos de Diferenciación Mielomonocítica/genética , Astrocitos/patología , Astrocitos/efectos de la radiación , Encéfalo/patología , Núcleo Celular/patología , Núcleo Celular/efectos de la radiación , Cerebelo/patología , Cerebelo/efectos de la radiación , Citoplasma/patología , Citoplasma/efectos de la radiación , Giro Dentado/patología , Giro Dentado/efectos de la radiación , Lóbulo Frontal/patología , Lóbulo Frontal/efectos de la radiación , Expresión Génica/efectos de la radiación , Hipocampo/patología , Hipocampo/efectos de la radiación , Masculino , Neuronas/patología , Neuronas/efectos de la radiación , Lóbulo Parietal/patología , Lóbulo Parietal/efectos de la radiación , Reacción en Cadena de la Polimerasa , Traumatismos Experimentales por Radiación/patología , Ratas , Ratas Sprague-Dawley
13.
Brain Res ; 1094(1): 207-16, 2006 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-16762327

RESUMEN

Several clinical and experimental reports suggest that low-dose irradiation of an established epileptic focus can reduce the occurrence of spontaneous seizures. Conversely, some recent reports suggest that under some conditions low-dose irradiation may have disinhibitory effects on seizure expression. Here, we have investigated mechanistic aspects of this phenomenon in the kindling model of epilepsy by applying focal irradiation at various points during kindling development. Rats were kindled to stage 5 by afterdischarge-threshold electrostimulation of the left amygdala. Treatment groups were irradiated using a collimated X-ray beam (18 MV) either prior to kindling, at kindling stage 3, or at kindling stage 5, by exposure of the left amygdala to a single-fraction central-axis dose of 25 Gy. Generalized seizure thresholds (GSTs) were subsequently assayed at weekly intervals for 10 weeks and at monthly intervals for an additional 3 months, along with the severity of the evoked seizures. Irradiation produced no significant effects on seizure threshold, but did produce persistent changes in seizure severity which varied as a function of the timing of irradiation. Relative to sham irradiated controls, the occurrence of stage 6 seizures was significantly increased by irradiation prior to kindling, but was unaffected by irradiation at kindling stage 3, and significantly reduced by irradiation at kindling stage 5. Quantitative immunohistochemical assays for neuron and astrocyte densities within the amygdala and hippocampus revealed only subtle changes in neuronal density within the dentate granule cell layer. These results are discussed in relation to mechanisms of seizure- and radiation-induced plasticity.


Asunto(s)
Amígdala del Cerebelo/efectos de la radiación , Epilepsia/radioterapia , Hipocampo/efectos de la radiación , Excitación Neurológica/efectos de la radiación , Vías Nerviosas/efectos de la radiación , Amígdala del Cerebelo/patología , Amígdala del Cerebelo/fisiopatología , Animales , Astrocitos/patología , Astrocitos/efectos de la radiación , Biomarcadores/metabolismo , Recuento de Células , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Estimulación Eléctrica , Epilepsia/fisiopatología , Epilepsia/prevención & control , Proteína Ácida Fibrilar de la Glía/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Excitación Neurológica/fisiología , Masculino , Proteínas del Tejido Nervioso/metabolismo , Vías Nerviosas/patología , Vías Nerviosas/fisiopatología , Plasticidad Neuronal/fisiología , Plasticidad Neuronal/efectos de la radiación , Neuronas/patología , Neuronas/efectos de la radiación , Proteínas Nucleares/metabolismo , Dosis de Radiación , Radiación Ionizante , Radioterapia/métodos , Ratas , Ratas Wistar , Factores de Tiempo , Resultado del Tratamiento
14.
Cancer Biol Ther ; 3(8): 739-51, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15197348

RESUMEN

We examined the impact of purified bacterially synthesized GST-MDA-7 (IL-24) and ionizing radiation on the proliferation and survival of nonestablished human glioblastoma multiforme (GBM) cells. Glioma cell types expressing mutated PTEN and p53 molecules, activated ERBB1VIII, overexpressing wild type ERBB1 or without receptor overexpression were selected. In MTT assays, GST-MDA-7 caused a dose-dependent reduction in the proliferation of nonestablished glioma cells; however only at higher concentrations did GST-MDA-7 reduce cell viability. The anti-proliferative and cytotoxic effects of GST-MDA-7 were enhanced by radiation in a greater than additive fashion that correlated with JNK1/2/3 activation. The reduction in cell growth and enhancement in cell killing by the combination of GST-MDA-7 and radiation were blocked by an ROS scavenger, N-acetyl cysteine (NAC), a JNK1/2/3 inhibitor SP600125, a pan-caspase inhibitor (zVAD) and by an inhibitor of caspase 9 (LEHD), but not by an inhibitor of caspase 8 (IETD). Low concentrations of either GST-MDA-7 or radiation reduced clonogenic survival, however colony formation ability was significantly further decreased when the two treatments were combined, which was also blocked by inhibition of caspase 9 function. In general agreement with activation of the intrinsic caspase pathway, cell death correlated with reduced BCL-XL expression and with increased levels of the pro-apoptotic proteins BAD and BAX. Inhibition of caspase 9 after combination treatment blunted neither JNK1/2/3 activation nor the enhanced expression of BAD and BAX, but did block caspase 3 cleavage, reduced expression of BCL-XL and inhibition of ERK1/2 activity. In contrast, incubation with NAC blocked JNK1/2/3 activation and cell killing, but not the increases in BAD and BAX expression. These findings argue that after combination treatment JNK1/2/3 activation is a primary pro-apoptotic event and loss of BCL-XL expression and ERK1/2 activity are secondary caspase-dependent processes. This data also argues that GST- MDA-7 induces two parallel pro-apoptotic pathways via ROS-dependent and -independent mechanisms. Infection of primary human astrocytes with a recombinant adenovirus to express MDA-7, Ad.mda-7, but not infection with either Ad.cmv or Ad.mda-7SP- lacking MDA-7 secretion, resulted in the suppression of GBM cell colony formation in soft agar overlay assays, an effect that was enhanced in a greater than additive fashion by radiation. Collectively, our findings demonstrate that MDA-7 reduces proliferation and enhances the radiosensitivity of nonestablished human GBM cells in vitro, and when grown in 3 dimensions, and that sensitization occurs independently of basal EGFR/ERK1/2/AKT activity or the functions of PTEN and p53.


Asunto(s)
Apoptosis/efectos de la radiación , Neoplasias Encefálicas/terapia , Proliferación Celular/efectos de la radiación , Glioblastoma/terapia , Interleucinas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Acetilcisteína/farmacología , Adenoviridae/genética , Adyuvantes Inmunológicos/farmacología , Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/efectos de la radiación , Neoplasias Encefálicas/metabolismo , Caspasa 8 , Caspasa 9 , Inhibidores de Caspasas , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Receptores ErbB/metabolismo , Genes Supresores de Tumor , Genes erbB-1/fisiología , Glioblastoma/metabolismo , Humanos , Interleucinas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , MAP Quinasa Quinasa 4 , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosfohidrolasa PTEN , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Tolerancia a Radiación , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteína X Asociada a bcl-2 , Proteína bcl-X
15.
J Biol Chem ; 275(25): 19275-81, 2000 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-10858458

RESUMEN

Expression of the src homology 3 (SH3) domain-containing expressed in tumorigenic astrocytes (SETA) gene is associated with the tumorigenic state in astrocytes. SETA encodes a variety of adapter proteins containing either one or two SH3 domains, as suggested by the sequence heterogeneity of isolated cDNAs. Using both SH3 domains in a yeast two-hybrid screen of a glial progenitor cell cDNA library, we isolated the rat homolog of the ALG-2-interacting protein 1 or ALG-2-interacting protein X (AIP1/Alix). In vitro confrontation experiments showed that the SH3-N domain of SETA interacted with the proline-rich C terminus of AIP1. In co-immunoprecipitation experiments, SETA and AIP1 interacted and could form a complex with apoptosis-linked gene 2 protein. Endogenous SETA and AIP1 proteins showed similar patterns of staining in primary rat astrocytes. Misexpression of a variety of SETA protein isoforms in these astrocytes revealed that they localized to the actin cytoskeleton. Furthermore, SETA proteins containing the SH3-N domain were able to sensitize astrocytes to apoptosis induced by UV irradiation. Expression of the isolated SH3-N domain had the greatest effect in these experiments, indicating that interference in the interaction between endogenous SETA and AIP1 sensitizes astrocytes to apoptosis in response to DNA damage.


Asunto(s)
Apoptosis/fisiología , Astrocitos/citología , Proteínas de Unión al Calcio/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Neoplasias/fisiología , Proteínas del Tejido Nervioso/fisiología , Actinas/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Reguladoras de la Apoptosis , Astrocitos/efectos de la radiación , ADN Complementario , Datos de Secuencia Molecular , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Unión Proteica , Ratas , Homología de Secuencia de Aminoácido , Técnicas del Sistema de Dos Híbridos , Rayos Ultravioleta , Xenopus , Dominios Homologos src
16.
Br J Cancer ; 77(10): 1612-20, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9635836

RESUMEN

This study compares the effect of gamma-linolenic acid (GLA) and its precursor linoleic acid (LA) on survival of 36B10 malignant rat astrocytoma cells and 'normal' rat astrocytes. GLA was cytotoxic to 36B10 cells but not to astrocytes. By contrast, LA supplementation did not affect the survival of either cell types. There were minor differences in the uptake, distribution and use of radiolabelled GLA and LA by the 36B10 cells and astrocytes. GLA and LA supplementation increased the total polyunsaturated fatty acid (PUFA) content of the cells indicating increased oxidative potential. However, elevated levels of 8-isoprostane, an indicator of increased oxidative stress, were only observed in the GLA supplemented 36B10 cells. Addition of the antioxidant trolox to GLA-enriched 36B10 cells blocked the cytotoxic effect. Further, GLA enhanced the radiation sensitivity of the astrocytoma cells but not the astrocytes; trolox blocked the GLA-mediated increase in astrocytoma cell radiosensitivity. LA did not affect the radiation response of either cell type. While cyclo-oxygenase inhibitors did not affect GLA cytotoxicity, they blocked the enhanced radiation response of GLA-supplemented cells. The lipoxygenase inhibitor NDGA did not affect the toxicity produced by GLA. Thus, GLA is toxic to the neoplastic astrocytoma cells but not to normal astrocytes.


Asunto(s)
Astrocitos/efectos de los fármacos , Astrocitoma/tratamiento farmacológico , Neoplasias Encefálicas/tratamiento farmacológico , Ácido alfa-Linolénico/farmacología , Ácido gammalinolénico/farmacología , Animales , Antineoplásicos/farmacología , Antioxidantes/farmacología , Astrocitos/metabolismo , Astrocitos/efectos de la radiación , Astrocitoma/metabolismo , Astrocitoma/radioterapia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/radioterapia , Muerte Celular , Cromanos/farmacología , Inhibidores de la Ciclooxigenasa/farmacología , Dinoprost/análogos & derivados , Dinoprost/análisis , F2-Isoprostanos , Ibuprofeno/farmacología , Indometacina/farmacología , Ratas , Ratas Sprague-Dawley , Células Tumorales Cultivadas , Vitamina E/administración & dosificación , Ácido alfa-Linolénico/metabolismo , Ácido gammalinolénico/metabolismo
17.
Radiat Res ; 143(1): 1-7, 1995 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7597135

RESUMEN

Radiation-induced gene expression was examined in rat astrocyte cultures using differential display of mRNA via reverse transcriptase-polymerase chain reaction. A 0.3-kb cDNA that was consistently observed in irradiated cultures but not in unirradiated cultures was cloned and sequenced. It was found to be identical to Ptk-3, a receptor tyrosine kinase gene identified recently. The protein encoded by Ptk-3 is a member of a novel class of receptor tyrosine kinases whose extracellular domain contains regions of homology with coagulation factors V and VIII and complement component C1. Northern blot analysis revealed that the expression of Ptk-3 was increased in rat astrocytes by 0.5 h after exposure to 10 Gy and remained at the same elevated level for at least 24 h. The maximum increase occurred after 5 Gy. Cloning studies indicated the presence of at least two Ptk-3 mRNA transcripts, which are probably the result of an alternative splicing mechanism. The short isoform lacks a 37-amino acid sequence in the glycine/proline-rich juxtamembrane region. The splicing pattern of the Ptk-3 gene was not altered by radiation. However, the ratios of the longer to the shorter mRNA transcripts differed between adult cortex, neonatal cortex and in vitro astrocyte cultures.


Asunto(s)
Astrocitos/efectos de la radiación , Regulación Enzimológica de la Expresión Génica/efectos de la radiación , Proteínas Tirosina Quinasas Receptoras/genética , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Astrocitos/enzimología , Secuencia de Bases , Northern Blotting , Células Cultivadas , Clonación Molecular , Cartilla de ADN , ADN Complementario , Receptor con Dominio Discoidina 1 , Datos de Secuencia Molecular , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley
18.
Stereotact Funct Neurosurg ; 61(3): 118-28, 1993.
Artículo en Inglés | MEDLINE | ID: mdl-7800977

RESUMEN

In order to observe the postoperative focal changes caused by radiofrequency lesions in the ventrolateral nucleus of the thalamus in stereotactic treatment of Parkinson's disease, similar lesions were produced in cat. The CT features and their correlative pathologic changes at different intervals were divided into three types, four stages and three zones. The degree of susceptibility of nervous tissue to radiofrequency, repair characteristics and the mechanism of CT enhancement are discussed in light of the pathologic changes.


Asunto(s)
Ondas de Radio , Tálamo/efectos de la radiación , Animales , Astrocitos/patología , Astrocitos/efectos de la radiación , Capilares/patología , Capilares/efectos de la radiación , Gatos , Eritrocitos/patología , Eritrocitos/efectos de la radiación , Masculino , Microglía/patología , Microglía/efectos de la radiación , Tálamo/diagnóstico por imagen , Tálamo/patología , Tomografía Computarizada por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA