Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Anat ; 230(2): 197-202, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27761896

RESUMEN

Transitory cavities associated with the ventricular system represent probably one of the most unique features in the developing mammalian brain. In rodents, the cavities exist transiently in the developing brain and do not appear to be associated with any pathological events. Among the various cavities, the pyramidal-shaped cavum septum pellucidum (CSP) located beneath the corpus callosum and between the lateral ventricles is most well defined. In addition to the CSP are the bilateral subependymal cysts that are consistently associated with the third and fourth ventricles as well as the aqueduct. The cavities/cysts contain a large number of amoeboid microglia expressing surface receptors and hydrolytic enzymes common to tissue macrophages. The significance of these cavities in the developing brain remains a conjecture. Firstly, the cavity walls are free of an apparent epithelial lining; hence, it is speculated that the cavities that appear to communicate with the widened neighboring interstitial tissue spaces may have resulted from physical traction due to the rapid growth of the perinatal brain. Secondly, the cavities contain prominent clusters of amoeboid microglia that may be involved in clearing the debris of degenerating axons and cells resulting from the early brain tissue remodeling. With the increase in brain tissue compactness following the beginning of myelination in the second postnatal week, all cavities are obliterated; concomitantly, the number of amoeboid microglia in them diminishes and all this might signal further maturation of the brain.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Ventrículos Cerebrales/crecimiento & desarrollo , Quistes , Animales , Encéfalo/ultraestructura , Ventrículos Cerebrales/ultraestructura , Cuerpo Calloso/crecimiento & desarrollo , Cuerpo Calloso/ultraestructura , Quistes/ultraestructura , Humanos
2.
Neural Plast ; 2017: 7351238, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28744378

RESUMEN

Spinal cord injury (SCI) often results in death of spinal neurons and atrophy of muscles which they govern. Thus, following SCI, reorganizing the lumbar spinal sensorimotor pathways is crucial to alleviate muscle atrophy. Tail nerve electrical stimulation (TANES) has been shown to activate the central pattern generator (CPG) and improve the locomotion recovery of spinal contused rats. Electroacupuncture (EA) is a traditional Chinese medical practice which has been proven to have a neural protective effect. Here, we examined the effects of TANES and EA on lumbar motor neurons and hindlimb muscle in spinal transected rats, respectively. From the third day postsurgery, rats in the TANES group were treated 5 times a week and those in the EA group were treated once every other day. Four weeks later, both TANES and EA showed a significant impact in promoting survival of lumbar motor neurons and expression of choline acetyltransferase (ChAT) and ameliorating atrophy of hindlimb muscle after SCI. Meanwhile, the expression of neurotrophin-3 (NT-3) in the same spinal cord segment was significantly increased. These findings suggest that TANES and EA can augment the expression of NT-3 in the lumbar spinal cord that appears to protect the motor neurons as well as alleviate muscle atrophy.


Asunto(s)
Neuronas Motoras/patología , Neuronas Motoras/fisiología , Músculo Esquelético/patología , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Cola (estructura animal)/inervación , Animales , Células del Asta Anterior/metabolismo , Células del Asta Anterior/patología , Células del Asta Anterior/fisiología , Estimulación Eléctrica , Electroacupuntura , Femenino , Neuronas Motoras/metabolismo , Atrofia Muscular , Neurotrofina 3/metabolismo , Ratas Sprague-Dawley , Médula Espinal , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/terapia
3.
J Neuroinflammation ; 12: 11, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25600517

RESUMEN

BACKGROUND: Activated microglial cells release an excess of inflammatory mediators after an ischemic stroke. We reported previously that scutellarin effectively suppressed the inflammatory response induced by activated microglia in rats subjected to middle cerebral artery occlusion (MCAO); however, the mechanism via which scutellarin exerts its effects on microglial activation has not been explored. This study aimed to elucidate if scutellarin can regulate the Notch pathway that is linked to microglia activation in MCAO rat, and in lipopolysaccharide (LPS)-induced BV-2 microglia. Along with this, we also investigated some characteristic behavioral responses of activated microglia. METHODS: Expression of various members of the Notch pathway, including Notch-1, intracellular Notch receptor domain (NICD), recombining binding protein suppressor of hairless (RBP-JK) and transcription factor hairy and enhancer of split-1 (Hes-1) in activated microglia was assessed by immunofluorescence staining and western blot after experimental MCAO and in vitro LPS activation. The effect of scutellarin on migration of microglia was determined by the transwell chamber assay as well as expression of monocyte chemoattractant protein-1 (MCP-1). The morphological change of microglia induced by scutellarin was detected by F-actin staining and electron microscopy. RESULTS: Scutellarin markedly attenuated the expression of NF-κB, Notch-1, NICD, RBP-JK and Hes-1 both in vivo and in activated microglia. It decreased the expression of MCP-1 and microglial migration, but increased the ability of microglia adhesion. Scutellarin also altered the phenotype of microglia by causing rearrangement or reorganization of its cytoskeleton. CONCLUSIONS: The results suggest that scutellarin regulates the activation of microglia via the Notch pathway and concurrently induces morphological and functional changes in activated microglia.


Asunto(s)
Apigenina/uso terapéutico , Movimiento Celular/efectos de los fármacos , Glucuronatos/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos , Actinas/metabolismo , Animales , Apigenina/farmacología , Adhesión Celular/efectos de los fármacos , Línea Celular Transformada , Cerebro/efectos de los fármacos , Cerebro/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Glucuronatos/farmacología , Lipopolisacáridos/farmacología , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/ultraestructura , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas de Unión al GTP rho/metabolismo
4.
BMC Neurosci ; 16: 84, 2015 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-26608466

RESUMEN

BACKGROUND: Scutellarin, an anti-inflammatory agent, effectively suppressed microglia activation in rats with middle cerebral artery occlusion (MCAO). Robust microglia activation, acute in onset, was followed by astrogliosis. This study was aimed to determine if scutellarin would also affect the reactive astrocytes that play an important role in tissue repair. Expression of GFAP and Notch-1 and its members: Notch receptor intracellular domain (NICD), and transcription factor hairy and enhancer of split-1 (HES-1), together with nestin and proinflammatory mediators was assessed by immunofluorescence staining in TNC1 astrocytes treated, respectively, with BV-2 conditioned medium (CM) and CM + lipopolysaccharide (LPS) (CM + L) serving as the controls, and conditioned medium derived from LPS-activated BV-2 cells pretreated with scutellarin (CM + SL). Study of the above biomarkers was then extended to reactive astrocytes in scutellarin injected MCAO rats. RESULTS: TNC1 astrocytes remained relatively unreactive in terms of expression of different biomarkers to direct scutellarin treatment when compared with the control cells. In comparison to cells in the control medium (CM, CM + L), they responded vigorously to CM + SL as evidenced by the enhanced protein expression of GFAP, Notch-1, NICD and HES-1 coupled with that of nestin, TNF-α, IL-1ß, and iNOS by Western and immunofluorescence analysis. Electron microscopy showed marked hypertrophy and cell expansion of TNC1 astrocytes bearing many filamentous processes indicative of enhanced astrocyte reaction when treated with CM + SL. In MCAO rats, scutellarin also augmented the expression of the above markers in reactive astrocytes; moreover, astrocytes were evidently hypertrophic. CONCLUSIONS: The results suggest that scutellarin regulates astrogliosis; more importantly, it is microglia-mediated as demonstrated in vitro. Increased expression of Notch signaling in synchrony with nestin may be linked to proliferation and "de-differentiation" of reactive astrocytes; the significance of enhanced TNF-α, IL-1ß and iNOS expression in reactive astrocytes by scutellarin may be neuroprotective but this remains speculative.


Asunto(s)
Apigenina/farmacología , Astrocitos/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Gliosis/tratamiento farmacológico , Glucuronatos/farmacología , Microglía/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Antiinflamatorios/farmacología , Astrocitos/patología , Astrocitos/fisiología , Isquemia Encefálica/complicaciones , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Gliosis/etiología , Gliosis/patología , Gliosis/fisiopatología , Infarto de la Arteria Cerebral Media , Masculino , Ratones , Microglía/patología , Microglía/fisiología , Ratas Sprague-Dawley
5.
NMR Biomed ; 28(9): 1069-77, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26152641

RESUMEN

Blast-induced traumatic brain injury is on the rise, predominantly as a result of the use of improvised explosive devices, resulting in undesirable neuropsychological dysfunctions, as demonstrated in both animals and humans. This study investigated the effect of open-field blast injury on the rat brain using multi-echo, susceptibility-weighted imaging (SWI). Multi-echo SWI provided phase maps with better signal-to-noise ratio (SNR) and contrast-to-noise ratio (CNR), making it a sensitive technique for brain injury. Male Sprague-Dawley rats were subjected to a survivable blast of 180 kPa. The visibility of blood vessels of varying sizes improved with multi-echo SWI. Reduced signal intensity from major vessels post-blast indicates increased deoxyhaemoglobin. Relative cerebral blood flow was computed from filtered phase SWI images using inferred changes in oxygen saturation from major blood vessels. Cerebral blood flow decreased significantly at day 3 and day 5 post-blast compared with that pre-blast. This was substantiated by the upregulation of ß-amyloid precursor protein (ß-APP), a marker of ischaemia, in the neuronal perikaya of the cerebral cortex, as observed by immunofluorescence, and in the cortical tissue by western blot analysis. Our findings indicate the presence of brain ischaemia in post-blast acute phase of injury with possible recovery subsequently. Our results from cerebrovascular imaging, histology and staining provide an insight into the ischaemic state of the brain post-blast and may be useful for prognosis and outcome.


Asunto(s)
Traumatismos por Explosión/patología , Lesiones Encefálicas/patología , Imagen por Resonancia Magnética/métodos , Precursor de Proteína beta-Amiloide/análisis , Animales , Circulación Cerebrovascular , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Sprague-Dawley , Relación Señal-Ruido
6.
Exp Eye Res ; 130: 73-86, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25433125

RESUMEN

This study was carried out to investigate the roles of tight junction (TJ) proteins and other factors in the increased permeability of the blood retinal barrier (BRB) affecting the immature neonatal retina following a hypoxic insult. The expression of endothelial TJ proteins such as claudin-5, occludin and zonula occludens-1 (ZO-1) and endothelial cell specific molecule-1 (ESM-1), and associated structural changes in the blood vessels were analyzed in the retinas of 1-day-old Wistar rats subjected to hypoxia for 2 h and subsequently sacrificed at different time points ranging from 3 h to 14 d. The mRNA and protein expression of claudin-5, occludin & ZO-1 was found to be reduced in the hypoxic retina, although, at the ultrastructural level, the TJ between the endothelial cells and retinal pigment epithelial cells appeared to be intact. Following the hypoxic insult vascular endothelial cells frequently showed presence of cytoplasmic vacuoles, vacuolated mitochondria and multivesicular aggregations projecting into the lumen of the capillaries. The expression of ESM-1 in the immature retinas was found to be increased following hypoxic exposure. The structural and molecular changes in the hypoxic neonatal retinas were consistent with a hypoxia induced impairment of the BRB. Hypoxia reduced the expression of TJ proteins in the neonatal retina, but the role of increased ESM-1 expression in this process warrants further investigation.


Asunto(s)
Claudina-5/genética , Endotelio Vascular/ultraestructura , Hipoxia/patología , Ocludina/genética , Retina/crecimiento & desarrollo , Vasos Retinianos/ultraestructura , Proteína de la Zonula Occludens-1/genética , Animales , Animales Recién Nacidos , Barrera Hematorretinal , Western Blotting , Permeabilidad Capilar , Claudina-5/metabolismo , Endotelio Vascular/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Microscopía Electrónica , Ocludina/metabolismo , Proteoglicanos/genética , Proteoglicanos/metabolismo , ARN Mensajero/genética , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/ultraestructura , Vasos Retinianos/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
7.
J Neuroinflammation ; 11: 176, 2014 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-25297558

RESUMEN

BACKGROUND: The damaging effect of combustion smoke inhalation on the lung is widely reported but information on its effects on the olfactory bulb is lacking. This study sought to determine the effects of smoke inhalation on the olfactory bulb, whose afferent input neurons in the nasal mucosa are directly exposed to external stimuli, such as smoke. METHODS: Adult male Sprague-Dawley rats were subjected to combustion smoke inhalation and sacrificed at different time points. Changes in olfactory bulb proteins including vascular endothelial growth factor (VEGF), inducible nitric oxide synthase (iNOS), endothelial nitric oxide synthase (eNOS), neuronal nitric oxide synthase (nNOS), Na+-K+-Cl- cotransporter 1 (NKCC1), glial fibrillary acidic protein (GFAP), and aquaporin-4 (AQP4) were evaluated by Western blot analysis. In addition, ELISA was conducted for cytokine and chemokine levels, and double immunofluorescence labeling was carried out for GFAP/VEGF, GFAP/AQP4, NeuN/nNOS, GFAP/NKCC1, NeuN/NKCC1, GFAP/Rhodamine isothiocyanate (RITC), and transferase dUTP nick end labeling (TUNEL). Aminoguanidine was administered to determine the effects of iNOS inhibition on the targets probed after smoke inhalation. RESULTS: The results showed a significant increase in VEGF, iNOS, eNOS, nNOS, NKCC1, and GFAP expression in the bulb tissues, with corresponding increases in inflammatory cytokines and chemokines after smoke inhalation. Concurrent to this was a drastic increase in AQP4 expression and RITC permeability. Aminoguanidine administration decreased the expression of iNOS and RITC extravasation after smoke inhalation. This was coupled with a significant reduction in incidence of TUNEL + cells that was not altered with administration of L-NG-nitroarginine methyl ester (L-NAME). CONCLUSIONS: These findings suggest that the upregulation of iNOS in response to smoke inhalation plays a major role in the olfactory bulb inflammatory pathophysiology, along with a concomitant increase in pro-inflammatory molecules, vascular permeability, and edema. Overall, these findings indicate that the olfactory bulb is vulnerable to smoke inhalation.


Asunto(s)
Inflamación/patología , Bulbo Olfatorio/patología , Humo/efectos adversos , Animales , Apoptosis , Western Blotting , Citocinas/análisis , Técnica del Anticuerpo Fluorescente , Inflamación/metabolismo , Masculino , Bulbo Olfatorio/metabolismo , Ratas , Ratas Sprague-Dawley
8.
J Neuroinflammation ; 11: 148, 2014 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-25148928

RESUMEN

BACKGROUND: We reported previously that amoeboid microglial cells in the postnatal rat brain expressed 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase) both in vivo and in vitro; however, the functional role of CNPase in microglia has remained uncertain. This study extended the investigation to determine CNPase expression in activated microglia derived from cell culture and animal models of brain injury with the objective to clarify its putative functions. METHODS: Three-day-old Wistar rats were given an intraperitoneal injection of lipopolysaccharide to induce microglial activation, and the rats were killed at different time points. Along with this, primary cultured microglial cells were subjected to lipopolysaccharide treatment, and expression of CNPase was analyzed by real-time reverse transcription PCR and immunofluorescence. Additionally, siRNA transfection was employed to downregulate CNPase in BV-2 cells. Following this, inducible nitric oxide synthase, IL-1ß and TNF-α were determined at mRNA and protein levels. Reactive oxygen species and nitric oxide were also assessed by flow cytometry and colorimetric assay, respectively. In parallel to this, CNPase expression in activated microglia was also investigated in adult rats subjected to fluid percussion injury as well as middle cerebral artery occlusion. RESULTS: In vivo, CNPase immunofluorescence in activated microglia was markedly enhanced after lipopolysaccharide treatment. A similar feature was observed in the rat brain after fluid percussion injury and middle cerebral artery occlusion. In vitro, CNPase protein and mRNA expression was increased in primary microglia with lipopolysaccharide stimulation. Remarkably, inducible nitric oxide synthase, IL-1ß, TNF-α, reactive oxygen species and nitric oxide were significantly upregulated in activated BV-2 cells with CNPase knockdown. siRNA knockdown of CNPase increased microglia migration; on the other hand, microglial cells appeared to be arrested at G1 phase. CONCLUSIONS: The present results have provided the first morphological and molecular evidence that CNPase expression is increased in activated microglia. CNPase knockdown resulted in increased expression of various inflammatory mediators. It is concluded that CNPase may play an important role as a putative anti-inflammatory gene both in normal and injured brain.


Asunto(s)
2',3'-Nucleótido Cíclico Fosfodiesterasas/metabolismo , Lesiones Encefálicas/patología , Regulación Enzimológica de la Expresión Génica/fisiología , Microglía/enzimología , Animales , Animales Recién Nacidos , Lesiones Encefálicas/etiología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Lipopolisacáridos/farmacología , Ratones , Microglía/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Accidente Cerebrovascular/complicaciones , Factores de Tiempo
9.
BMC Neurosci ; 15: 125, 2014 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-25416145

RESUMEN

BACKGROUND: In response to cerebral ischemia, activated microglia release excessive inflammatory mediators which contribute to neuronal damage. Therefore, inhibition of microglial over-activation could be a therapeutic strategy to alleviate various microglia-mediated neuroinflammation. This study was aimed to elucidate the anti-inflammatory effects of Scutellarin and Edaravone given either singly, or in combination in activated microglia in rats subjected to middle cerebral artery occlusion (MCAO), and in lipopolysaccharide (LPS)-induced BV-2 microglia. Expression of proinflammatory cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1ß), and inducible nitric oxide synthase (iNOS) was assessed by immunofluorescence staining and Western blot. Reactive oxygen species (ROS) and nitric oxide (NO) levels were determined by flow cytometry and fluorescence microscopy, respectively. RESULTS: In vivo, both Edaravone and Scutellarin markedly reduced the infarct cerebral tissue area with the latter drug being more effective with the dosage used; furthermore, when used in combination the reduction was more substantial. Remarkably, a greater diminution in distribution of activated microglia was observed with the combined drug treatment which also attenuated the immunoexpression of TNF-α, IL-1ß and iNOS to a greater extent as compared to the drugs given separately. In vitro, both drugs suppressed upregulated expression of inflammatory cytokines, iNOS, NO and ROS in LPS-induced BV-2 cells. Furthermore, Edaravone and Scutellarin in combination cumulatively diminished the expression levels of the inflammatory mediators being most pronounced for TNF-α as evidenced by Western blot. CONCLUSION: The results suggest that Edaravone and Scutellarin effectively suppressed the inflammatory responses in activated microglia, with Scutellarin being more efficacious within the dosage range used. Moreover, when both drugs were used in combination, the infarct tissue area was reduced more extensively; also, microglia-mediated inflammatory mediators notably TNF-α expression was decreased cumulatively.


Asunto(s)
Antiinflamatorios/farmacología , Antipirina/análogos & derivados , Apigenina/farmacología , Isquemia Encefálica/tratamiento farmacológico , Glucuronatos/farmacología , Microglía/efectos de los fármacos , Animales , Antipirina/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/patología , Isquemia Encefálica/inmunología , Isquemia Encefálica/patología , Recuento de Células , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Edaravona , Infarto de la Arteria Cerebral Media , Lipopolisacáridos , Masculino , Microglía/inmunología , Microglía/patología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Distribución Aleatoria , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
10.
Mol Cell Neurosci ; 56: 342-54, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23859823

RESUMEN

Excess glutamate mediates damage to oligodendroglia, resulting in myelination disturbances characteristic of hypoxic periventricular white matter (PWM) damage. We sought to examine if hypoxia altered the expression of astroglial excitatory amino acid transporters (EAAT1, EAAT2 and EAAT3) in the PWM, and, if so, whether it activated astroglial N-methyl D-aspartate receptors (NMDAR) which might lead to apoptosis of oligodendroglia. EAAT expression in the PWM of neonatal rats was measured at different time points after hypoxic exposure; it was attenuated at 7 and 14 d following hypoxia. Hypoxia prevented the uptake of glutamate by astroglial EAATs causing increased levels of extracellular glutamate. Excess glutamate augmented the expression of functional astroglial NMDAR. Following hypoxia, an increase in gap junction proteins between astroglia and oligodendroglia aided in the spreading of NMDAR-mediated excitotoxic calcium signals into the latter cell type triggering its apoptosis. Hence, dysregulated glutamate homeostasis is believed to contribute to hypoxia-induced death of oligodendroglia leading to neonatal PWM damage.


Asunto(s)
Apoptosis , Astrocitos/metabolismo , Ácido Glutámico/metabolismo , Hipoxia Encefálica/metabolismo , Oligodendroglía/metabolismo , Animales , Hipoxia de la Célula , Células Cultivadas , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Conexinas/genética , Conexinas/metabolismo , Proteínas de Transporte de Glutamato en la Membrana Plasmática/genética , Proteínas de Transporte de Glutamato en la Membrana Plasmática/metabolismo , Homeostasis , Hipoxia Encefálica/patología , Oligodendroglía/patología , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo
11.
J Neuroinflammation ; 10: 23, 2013 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-23388509

RESUMEN

BACKGROUND: Hypoxia induces microglial activation which causes damage to the developing brain. Microglia derived inflammatory mediators may contribute to this process. Toll-like receptor 4 (TLR4) has been reported to induce microglial activation and cytokines production in brain injuries; however, its role in hypoxic injury remains uncertain. We investigate here TLR4 expression and its roles in neuroinflammation in neonatal rats following hypoxic injury. METHODS: One day old Wistar rats were subjected to hypoxia for 2 h. Primary cultured microglia and BV-2 cells were subjected to hypoxia for different durations. TLR4 expression in microglia was determined by RT-PCR, western blot and immunofluorescence staining. Small interfering RNA (siRNA) transfection and antibody neutralization were employed to downregulate TLR4 in BV-2 and primary culture. mRNA and protein expression of tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1ß) and inducible nitric oxide synthase (iNOS) was assessed. Reactive oxygen species (ROS), nitric oxide (NO) and NF-κB levels were determined by flow cytometry, colorimetric and ELISA assays respectively. Hypoxia-inducible factor-1 alpha (HIF-1α) mRNA and protein expression was quantified and where necessary, the protein expression was depleted by antibody neutralization. In vivo inhibition of TLR4 with CLI-095 injection was carried out followed by investigation of inflammatory mediators expression via double immunofluorescence staining. RESULTS: TLR4 immunofluorescence and protein expression in the corpus callosum and cerebellum in neonatal microglia were markedly enhanced post-hypoxia. In vitro, TLR4 protein expression was significantly increased in both primary microglia and BV-2 cells post-hypoxia. TLR4 neutralization in primary cultured microglia attenuated the hypoxia-induced expression of TNF-α, IL-1ß and iNOS. siRNA knockdown of TLR4 reduced hypoxia-induced upregulation of TNF-α, IL-1ß, iNOS, ROS and NO in BV-2 cells. TLR4 downregulation-mediated inhibition of inflammatory cytokines in primary microglia and BV-2 cells was accompanied by the suppression of NF-κB activation. Furthermore, HIF-1α antibody neutralization attenuated the increase of TLR4 expression in hypoxic BV-2 cells. TLR4 inhibition in vivo attenuated the immunoexpression of TNF-α, IL-1ß and iNOS on microglia post-hypoxia. CONCLUSION: Activated microglia TLR4 expression mediated neuroinflammation via a NF-κB signaling pathway in response to hypoxia. Hence, microglia TLR4 presents as a potential therapeutic target for neonatal hypoxia brain injuries.


Asunto(s)
Encéfalo/metabolismo , Hipoxia Encefálica/metabolismo , Mediadores de Inflamación/metabolismo , Microglía/metabolismo , Receptor Toll-Like 4/fisiología , Animales , Animales Recién Nacidos , Encéfalo/patología , Supervivencia Celular/fisiología , Células Cultivadas , Hipoxia Encefálica/patología , Mediadores de Inflamación/fisiología , Ratas , Ratas Wistar
12.
J Neuroinflammation ; 10: 79, 2013 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-23819902

RESUMEN

BACKGROUND: The effect of primary blast exposure on the brain is widely reported but its effects on the eye remains unclear. Here, we aim to examine the effects of primary blast exposure on the retina. METHODS: Adult male Sprague-Dawley rats were exposed to primary blast high and low injury and sacrificed at 24 h, 72 h, and 2 weeks post injury. The retina was subjected to western analysis for vascular endothelial growth factor (VEGF), aquaporin-4 (AQP4), glutamine synthethase (GS), inducible nitric oxide synthase (NOS), endothelial NOS, neuronal NOS and nestin expression; ELISA analysis for cytokines and chemokines; and immunofluorescence for glial fibrillary acidic protein (GFAP)/VEGF, GFAP/AQP4, GFAP/nestin, GS/AQP4, lectin/iNOS, and TUNEL. RESULTS: The retina showed a blast severity-dependent increase in VEGF, iNOS, eNOS, nNOS, and nestin expression with corresponding increases in inflammatory cytokines and chemokines. There was also increased AQP4 expression and retinal thickness after primary blast exposure that was severity-dependent. Finally, a significant increase in TUNEL+ and Caspase-3+ cells was observed. These changes were observed at 24 h post-injury and sustained up to 2 weeks post injury. CONCLUSIONS: Primary blast resulted in severity-dependent pathological changes in the retina, manifested by the increased expression of a variety of proteins involved in inflammation, edema, and apoptosis. These changes were observed immediately after blast exposure and sustained up to 2 weeks suggesting acute and chronic injury mechanisms. These changes were most obvious in the astrocytes and Müller cells and suggest important roles for these cells in retina pathophysiology after blast.


Asunto(s)
Traumatismos por Explosión/patología , Retina/patología , Animales , Apoptosis/fisiología , Acuaporina 4/biosíntesis , Traumatismos por Explosión/metabolismo , Western Blotting , Muerte Celular/fisiología , Quimiocinas/metabolismo , Citocinas/metabolismo , Sustancias Explosivas , Técnica del Anticuerpo Fluorescente Indirecta , Proteína Ácida Fibrilar de la Glía/biosíntesis , Ácido Glutámico/metabolismo , Proteínas de Filamentos Intermediarios/biosíntesis , Masculino , Proteínas del Tejido Nervioso/biosíntesis , Nestina , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Nitritos/metabolismo , Ratas , Ratas Sprague-Dawley , Retina/metabolismo , Trinitrotolueno
13.
J Pineal Res ; 54(2): 190-206, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23113620

RESUMEN

The purpose of this study was to determine whether melatonin treatment would mitigate retinal ganglion cell (RGC) death in the developing retina following a hypoxic insult. Lipid peroxidation (LPO), glutathione (GSH), tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) concentrations, expression of vascular endothelial growth factor receptors, Flt-1 and Flk-1, release of cytochrome c from mitochondria, and caspase-3 expression were examined in the retinas of 1-day-old rats at 3 hr to 14 days after a hypoxic exposure. The mRNA and protein expression of Flt-1 and Flk-1 and the tissue concentration of LPO, TNF-α, and IL-1ß were upregulated significantly after the hypoxic exposure, whereas the content of GSH was decreased significantly. RGC cultures also showed increased LPO and decreased GSH levels after hypoxic exposure but these effects were reversed in cells treated with melatonin. TNF-α and IL-1ß expression was specifically located on microglial cells, whereas Flt-1 and Flk-1 was limited to RGCs as confirmed by double immunofluorescence labeling. Cultures of hypoxic microglial cells treated with melatonin showed a significant reduction in the release of these cytokines as compared to untreated hypoxic cells. Hypoxia induced increase in the cytosolic cytochrome c and caspase-3 in RGCs was attenuated with melatonin treatment. The results suggest that, in hypoxic injuries, melatonin is neuroprotective to RGCs in the developing retina through its antioxidative, anti-inflammatory, and anti-apoptotic effects. Melatonin suppressed Flt-1 and Flk-1 expression in retinal blood vessels, which may result in reduced retinal vascular permeability and it also preserved mitochondrial function as shown by a reduction in cytochrome c leakage into the cytosol. The results may have therapeutic implications for the management of retinopathy of prematurity.


Asunto(s)
Melatonina/toxicidad , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/efectos de los fármacos , Animales , Western Blotting , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Glutatión/metabolismo , Interleucina-1beta/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Células Ganglionares de la Retina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
14.
J Pineal Res ; 55(2): 121-30, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23551342

RESUMEN

Hypoxia is a crucial factor in tumor aggressiveness and resistance to therapy, especially in glioblastoma. Our previous results have shown that melatonin exerts antimigratory and anti-invasive action in glioblastoma cells under normoxia. However, the effect of melatonin on migration and invasion of glioblastoma cells under hypoxic condition remains poorly understood. Here, we show that melatonin strongly reduced hypoxia-mediated invasion and migration of U251 and U87 glioblastoma cells. In addition, we found that melatonin significantly blocked HIF-1α protein expression and suppressed the expression of downstream target genes, matrix metalloproteinase 2 (MMP-2) and vascular endothelial growth factor (VEGF). Furthermore, melatonin destabilized hypoxia-induced HIF-1α protein via its antioxidant activity against ROS produced by glioblastoma cells in response to hypoxia. Along with this, HIF-1α silencing by small interfering RNA markedly inhibited glioblastoma cell migration and invasion, and this appeared to be associated with MMP-2 and VEGF under hypoxia. Taken together, our findings suggest that melatonin suppresses hypoxia-induced glioblastoma cell migration and invasion via inhibition of HIF-1α. Considering the fact that overexpression of the HIF-1α protein is often detected in glioblastoma multiforme, melatonin may prove to be a potent therapeutic agent for this tumor.


Asunto(s)
Glioblastoma/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Hipoxia/patología , Melatonina/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Técnicas de Silenciamiento del Gen , Glioblastoma/genética , Glioblastoma/patología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Biomaterials ; 299: 122161, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37236138

RESUMEN

We previously constructed a three-dimensional gelatin sponge (3D-GS) scaffold as a delivery vehicle for therapeutic cells and trophic factors in the treatment of spinal cord injury (SCI), and this study aimed to assess the biosafety and efficacy of the scaffold in a non-human primate SCI model. However, because it has only been tested in rodent and canine models, the biosafety and efficacy of the scaffold should ideally be assessed in a non-human primate SCI model before its use in the clinic. No adverse reactions were observed over 8 weeks following 3D-GS scaffold implantation into in a Macaca fascicularis with hemisected SCI. Scaffold implantation also did not add to neuroinflammatory or astroglial responses already present at the injured site, suggesting good biocompatibility. Notably, there was a significant reduction in α-smooth muscle actin (αSMA)-positive cells at the injury/implantation interface, leading to alleviation of fibrotic compression of the residual spinal cord tissue. The regenerating tissue in the scaffold showed numerous cells migrating into the implant secreting abundant extracellular matrix, resulting in a pro-regenerative microenvironment. Consequently, nerve fiber regeneration, myelination, vascularization, neurogenesis, and electrophysiological improvements were achieved. These results indicated that the 3D-GS scaffold had good histocompatibility and effectiveness in the structural repair of injured spinal cord tissue in a non-human primate and is suitable for use in the treatment of patients with SCI.


Asunto(s)
Gelatina , Traumatismos de la Médula Espinal , Animales , Perros , Gelatina/química , Andamios del Tejido/química , Traumatismos de la Médula Espinal/terapia , Regeneración Nerviosa/fisiología , Médula Espinal , Primates
16.
J Neurosci ; 31(49): 17982-95, 2011 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-22159112

RESUMEN

This study was aimed to examine the role of iron in causing periventricular white matter (PWM) damage following a hypoxic injury in the developing brain. Along with iron, the expression of iron regulatory proteins (IRPs) and transferrin receptor (TfR), which are involved in iron acquisition, was also examined in the PWM by subjecting 1-d-old Wistar rats to hypoxia. Apart from an increase in iron levels in PWM, Perls' iron staining showed an increase of intracellular iron in the preponderant amoeboid microglial cells (AMCs) in the tissue. In response to hypoxia, the protein levels of IRP1, IRP2, and TfR in PWM and AMCs were significantly increased. In primary microglial cultures, administration of iron chelator deferoxamine reduced the generation of iron-induced reactive oxygen and nitrogen species and proinflammatory cytokines such as tumor necrosis factor-α and interleukin-1ß. Primary oligodendrocytes treated with conditioned medium from hypoxic microglia exhibited reduced glutathione levels, increased lipid peroxidation, upregulated caspase-3 expression, and reduced proliferation. This was reversed to control levels on treatment with conditioned medium from deferoxamine treated hypoxic microglia; also, there was reduction in apoptosis of oligodendrocytes. The present results suggest that excess iron derived primarily from AMCs might be a mediator of oligodendrocyte cell death in PWM following hypoxia in the neonatal brain.


Asunto(s)
Citocinas/metabolismo , Hipoxia/patología , Proteínas Reguladoras del Hierro/metabolismo , Hierro/metabolismo , Microglía/metabolismo , Oligodendroglía/fisiología , Animales , Animales Recién Nacidos , Muerte Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Corteza Cerebral/citología , Deferoxamina/farmacología , Modelos Animales de Enfermedad , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Glutatión/metabolismo , Etiquetado Corte-Fin in Situ , Peroxidación de Lípido/efectos de los fármacos , Microglía/efectos de los fármacos , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Receptores de Transferrina/metabolismo , Sideróforos/farmacología
17.
J Neuroinflammation ; 9: 173, 2012 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-22799524

RESUMEN

CD137 (4-1BB, TNFRSF9), a member of the tumor necrosis factor (TNF) receptor family, is a potent T cell co-stimulatory molecule. CD137 ligand (CD137L) is expressed by antigen presenting cells (APC) as a transmembrane protein and transmits activating signals into APC. In this study we investigated the effects of CD137L signaling in microglia, the resident APC in the central nervous system. In vitro, the murine microglia cell lines BV-2 and N9, as well as primary murine microglia responded with activation as evidenced by adherence and secretion of proinflammatory cytokines, MMP-9, and soluble intercellular adhesion molecule (ICAM). CD137L signaling is also important for microglia activation in vivo, since CD137L-deficient mice exhibited profoundly less microglia activation during experimental autoimmune encephalomyelitis (EAE) which is a well-established murine model for neuroinflammation and human multiple sclerosis (MS). Also CD137 is expressed in the CNS of mice during EAE. Activated microglia has been reported to mediate the destruction of axonal myelin sheaths and cause the death of oligodendrocytes, the main pathogenic mechanisms in EAE and MS. Corresponding to the lower microglia activation there were also fewer apoptotic oligodendrocytes in the CNS of CD137L-deficient mice. In vitro co-culture confirmed that CD137L-activated microglia induces apoptosis in oligodendrocytes, and identified reactive oxygen species as the mechanism of apoptosis induction. These data demonstrate activating effects of CD137L signaling to microglia, and show for the first time that the CD137 receptor/ligand system may be a mediator of neuroinflammatory and neurodegenerative disease, by activating microglia which in turn kill oligodendrocytes.


Asunto(s)
Ligando 4-1BB/metabolismo , Apoptosis/fisiología , Microglía/metabolismo , Oligodendroglía/citología , Oligodendroglía/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular , Células Cultivadas , Técnicas de Cocultivo , Femenino , Ratones , Ratones Endogámicos C57BL
18.
BMC Neurosci ; 13: 64, 2012 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-22697290

RESUMEN

BACKGROUND: Microglia, the resident immune cells of the central nervous system (CNS), have two distinct phenotypes in the developing brain: amoeboid form, known to be amoeboid microglial cells (AMC) and ramified form, known to be ramified microglial cells (RMC). The AMC are characterized by being proliferative, phagocytic and migratory whereas the RMC are quiescent and exhibit a slow turnover rate. The AMC transform into RMC with advancing age, and this transformation is indicative of the gradual shift in the microglial functions. Both AMC and RMC respond to CNS inflammation, and they become hypertrophic when activated by trauma, infection or neurodegenerative stimuli. The molecular mechanisms and functional significance of morphological transformation of microglia during normal development and in disease conditions is not clear. It is hypothesized that AMC and RMC are functionally regulated by a specific set of genes encoding various signaling molecules and transcription factors. RESULTS: To address this, we carried out cDNA microarray analysis using lectin-labeled AMC and RMC isolated from frozen tissue sections of the corpus callosum of 5-day and 4-week old rat brain respectively, by laser capture microdissection. The global gene expression profiles of both microglial phenotypes were compared and the differentially expressed genes in AMC and RMC were clustered based on their functional annotations. This genome wide comparative analysis identified genes that are specific to AMC and RMC. CONCLUSIONS: The novel and specific molecules identified from the trancriptome explains the quiescent state functioning of microglia in its two distinct morphological states.


Asunto(s)
Cuerpo Calloso/citología , Regulación del Desarrollo de la Expresión Génica/fisiología , Microglía/clasificación , Microglía/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Diferenciación Celular/genética , Línea Celular Transformada , Proliferación Celular , Análisis por Conglomerados , Citocinas/genética , Citocinas/metabolismo , Citoesqueleto/genética , Citoesqueleto/metabolismo , Perfilación de la Expresión Génica , Proteína 2 Inhibidora de la Diferenciación/genética , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Captura por Microdisección con Láser , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Wistar , Células Madre , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Br J Nutr ; 107(2): 229-41, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21733316

RESUMEN

Diets rich in SFA have been implicated in Alzheimer's disease (AD). There is strong evidence to suggest that microglial activation augments the progression of AD. However, it remains uncertain whether SFA can initiate microglial activation and whether this response can cause neuronal death. Using the BV-2 microglial cell line and primary microglial culture, we showed that palmitic acid (PA) and stearic acid (SA) could activate microglia, as assessed by reactive morphological changes and significantly increased secretion of pro-inflammatory cytokines, NO and reactive oxygen species, which trigger primary neuronal death. In addition, the mRNA level of these pro-inflammatory mediators determined by RT-PCR was also increased by PA and SA. We further investigated the intracellular signalling mechanism underlying the release of pro-inflammatory mediators from PA-activated microglial cells. The present results showed that PA activated the phosphorylation and nuclear translocation of the p65 subunit of NF-κB. Furthermore, pyrrolidine dithiocarbamate, a NF-κB inhibitor, attenuated the production of pro-inflammatory mediators except for IL-6 in PA-stimulated microglia. Administration of anti-Toll-like receptor (TLR)4-neutralising antibody repressed PA-induced NF-κB activation and pro-inflammatory mediator production. In conclusion, the present in vitro study demonstrates that SFA could activate microglia and stimulate the TLR4/NF-κB pathway to trigger the production of pro-inflammatory mediators, which may contribute to neuronal death.


Asunto(s)
Microglía/metabolismo , Ácido Palmítico/efectos adversos , Transducción de Señal , Ácidos Esteáricos/efectos adversos , Receptor Toll-Like 4/metabolismo , Factor de Transcripción ReIA/metabolismo , Regulación hacia Arriba , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Genes Reporteros/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Microglía/efectos de los fármacos , Microglía/patología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/antagonistas & inhibidores , Factor de Transcripción ReIA/antagonistas & inhibidores , Factor de Transcripción ReIA/genética , Regulación hacia Arriba/efectos de los fármacos
20.
J Pineal Res ; 52(2): 253-64, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21929682

RESUMEN

Melatonin, an endogenously produced neurohormone secreted mainly by the pineal gland, has a variety of physiological functions and neuroprotective effects. Saturated fatty acids (SFAs) have been known to induce neurotoxicity and oxidative stress in central nervous system injuries and neurodegenerative pathologies. However, the effect of melatonin on SFAs-induced cytotoxicity in astroglial cells, if any, has remained to be explored. This study reports that in primary cultured astroglial cells, melatonin significantly attenuated palmitic acid (PA)-induced cytotoxicity in a concentration- and time-dependent manner. Additionally, melatonin effectively suppressed PA-induced reactive oxygen species generation and prevented PA-induced apoptosis whereby the rise in Bax/Bcl-2 ratio and caspase-3 activation in astroglial cells was inhibited. However, it did not appear to exert an obvious effect on PA-induced intracellular calcium overload. Luzindole, a nonselective melatonin receptor antagonist, attenuated melatonin's promotion effect of cell survival and Stat3 phosphorylation, indicating that melatonin exerts its protective property in astroglial cells, at least in part, through the activation of membrane receptors and then Stat3 signaling pathway. Finally, melatonin had an inhibitory effect on the pro-inflammatory cytokine gene expression. The results suggest that melatonin may be an effective cytoprotective agent against PA-based cytotoxicity through modulating cell survival and inflammatory response in astroglial cells.


Asunto(s)
Astrocitos/efectos de los fármacos , Melatonina/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Apoptosis/efectos de los fármacos , Astrocitos/metabolismo , Western Blotting , Supervivencia Celular/efectos de los fármacos , Citocinas/biosíntesis , Citoprotección/efectos de los fármacos , Inhibidores Enzimáticos/toxicidad , Expresión Génica/efectos de los fármacos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Técnicas In Vitro , Ratones , Ratones Endogámicos BALB C , Ácido Palmítico/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA