Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neurochem Res ; 37(7): 1578-83, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22446845

RESUMEN

Long-lasting siRNA-based down-regulation of gene of interest can be achieved by lentiviral-based expression vectors driving the production of short hairpin RNA (shRNA). We investigated an attractive therapeutic approach to target the expression of proinflammatory GMF by using lentiviral vector encoding GMF-specific shRNA to reduce GMF levels in the spinal cord and brain of mice. To determine the effect of GMF-shRNA on GMF protein levels, we performed quantitative ELISA analysis in brain and in thoracic, cervical and lumbar regions of spinal cord from mice followed by GMF-shRNA (G-shRNA) or control shRNA (C-shRNA) treatments. Our results show a marked reduction of GMF protein levels in brain and spinal cord of mice treated with GMF-shRNA compared to control shRNA treatment. Consistent with the GMF protein analysis, the immunohistochemical examination of the spinal cord sections of EAE mice treated with GMF-shRNA showed significantly reduced GMF-immunoreactivity. Thus, the down-regulation of GMF by GMF-shRNA was efficient and wide spread in CNS as evident by the significantly reduced levels of GMF protein in the brain and spinal cord of mice.


Asunto(s)
Encéfalo/metabolismo , Regulación hacia Abajo , Factor de Maduración de la Glia/metabolismo , Médula Espinal/metabolismo , Animales , Secuencia de Bases , Ensayo de Inmunoadsorción Enzimática , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño
2.
Neurochem Int ; 60(3): 215-9, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22226840

RESUMEN

The role of glia maturation factor (GMF) in myelin oligodendrocyte glycoprotein (MOG) 35-55 peptide-induced experimental autoimmune encephalomyelitis (EAE) was investigated using GMF-deficient (GMF-KO) mice. We demonstrate that GMF-KO mice were resistant to the MOG 35-55 peptide-induced EAE as compared to wild type (Wt) mice (two in eight versus 10 in 10). Next, we examined the effect of administration of recombinant human GMF (rGMF) on MOG 35-55 peptide-induced EAE in mice. Daily administration of rGMF, staring days 1-14, resulted in significant exacerbation of clinical symptoms. Following rGMF injections, both GMF-KO (six in eight) and Wt mice (eight in eight) developed severe EAE (maximal clinical score of 3.5-4.0) with high frequency. The histological examination revealed severe infiltration of inflammatory cells in the spinal cord of MOG-immunized Wt mice while the resistance to EAE in GMF-KO mice was characterized by the absence of inflammatory cells. Administration of rGMF in Wt mice and GMF-KO mice resulted in a significant increase in infiltrating cells in the spinal cord following MOG-immunizations. We also evaluated cytokines and chemokines production as parameters of severity of inflammation in the spinal cord of Wt versus GMF-KO mice with and without GMF-reconstitution following MOG-immunizations. Cytokines (TNF-α, IFN-γ, IL-1ß, IL-6) and chemokines (CCL2, CCL3, CXCL10, GM-CSF) production were significantly greater in Wt mice than in GMF-KO mice following MOG-immunization. Furthermore, the reconstitution experiment with rGMF showed that the administration of rGMF in both, Wt mice and GMF-KO mice produced significant increase in the GMF-mediated cytokine/chemokine production.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Factor de Maduración de la Glia/fisiología , Glicoproteínas/toxicidad , Fragmentos de Péptidos/toxicidad , Animales , Quimiocinas/metabolismo , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Factor de Maduración de la Glia/genética , Humanos , Inmunización , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito , Proteínas Recombinantes/farmacología , Médula Espinal/patología
3.
Brain Res ; 1373: 230-9, 2011 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-21146509

RESUMEN

Glia maturation factor (GMF), a protein primarily localized in the central nervous system (CNS) was isolated, sequenced and cloned in our laboratory. We previously demonstrated that GMF mediates the experimental autoimmune encephalomyelitis (EAE)-induced production of pro-inflammatory cytokines and chemokines in the central nervous system of mice. In the present study we show that immunization with myelin oligodendrocyte glycoprotein peptide 35-55 (MOG35-55) caused an early onset (days 7-9 post immunization) and severe EAE with a mean peak score of 3.5 ± 0.5 in mice. Neutralization of GMF with four injections of anti-GMF antibody 5 to 11 days post immunization delayed the time of onset (days 12-14 post immunization) and significantly reduced the severity of EAE (mean peak score of 1.5 ± 0.4). Consistent with these clinical scores, histological examination of the CNS of these mice revealed profound differences between GMF-antibody treated mice and isotype matched control-antibody treated mice. Histological analysis of the spinal cord and brain showed severe inflammation and demyelination in the control antibody-treated mice whereas significantly reduced inflammation and demyelination was detected in GMF-antibody-treated mice at days 8, 16, and 24 post immunization. The decreased incidence and reduced severity of EAE in GMF-antibody-treated mice was consistent with the significantly reduced expressions of pro-inflammatory cytokines and chemokines. Our overall results demonstrate that neutralization of endogenous GMF with an affinity purified GMF antibody significantly decreased the inflammation, severity and progression of immunization induced active, passive and relapsing-remitting EAE. Treatment of mice with isotype-matched control antibody did not have any effect on EAE. Taken together, these results demonstrate the critical role of GMF in EAE, and GMF antibody as a potent anti-inflammatory therapeutic agent for effectively suppressing EAE in mouse models of major types of multiple sclerosis (MS).


Asunto(s)
Anticuerpos/uso terapéutico , Encefalomielitis Autoinmune Experimental/complicaciones , Encefalomielitis Autoinmune Experimental/patología , Factor de Maduración de la Glia/inmunología , Inflamación/tratamiento farmacológico , Inflamación/etiología , Neuronas/efectos de los fármacos , Animales , Citocinas/genética , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/etiología , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Glicoproteínas/efectos adversos , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos/efectos adversos , Toxina del Pertussis/efectos adversos , ARN Mensajero/metabolismo , Factores de Tiempo
4.
Neurobiol Dis ; 40(3): 593-8, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20696246

RESUMEN

Glia maturation factor (GMF), a primarily CNS localized protein was discovered and characterized in our laboratory. We previously demonstrated that GMF is the upstream regulator for excessive production and release of proinflammatory cytokines/chemokines in brain cells leading to the destruction of oligodendrocytes, the myelin forming cells, and neurons. We also reported that mice lacking endogenous GMF (GMF-deficient, GMF-KO) were resistant to myelin oligodendrocyte glycoprotein peptide 35-55 (MOG(35-55)) induced EAE, since immunization induced only delayed EAE with diminished severity. In the present study we show that a replication-defective adenovirus-GMF construct caused expression of GMF in CNS of GMF-KO mice and reinstated MOG(35-55) induced early and severe EAE. Our results show that MOG(35-55) immunization caused only a muted EAE and inflammation/demyelination in mice lacking endogenous GMF. The diminished incidence of EAE in GMF-KO mice was consistent with the significantly reduced expressions of cytokines/chemokines. The muted severity of EAE in GMF-KO mice was restored to full blown levels upon reintroduction of GMF using an adeno-GMF-virus (Adv-GMF) vector. Consistent with the clinical findings, histological examination of the CNS of mice with EAE revealed profound differences between wild type (Wt), GMF-KO, and GMF-KO mice with re-introduced GMF (GMF-KO+Adv-GMF). Spinal cord sections from mice with EAE were analyzed for the infiltration of mononuclear cells (inflammation) and myelin loss (demyelination). In Wt mice, 40% of spinal cord quadrants were positive for demyelination and 45% of spinal cord quadrants were positive for inflammation at the peak of EAE. Drastically reduced infiltrates (15%) and demyelination (10%) were found in GMF-KO mice that developed reduced severity of EAE. Upon GMF reintroduction in GMF-KO mice, MOG(35-55) immunization caused extensive monocytes infiltration (48%) and demyelination (46%), similar to that observed in the immunized Wt mice. The levels of cytokine/chemokine in the spinal cords of mice at three time points, corresponding to the onset, peak severity and recovery period of EAE, show a distinct pattern of very large increases in IFN-γ, TNF-α, GM-CSF and MCP-1 in Wt and GMF-KO+Adv-GMF mice compared to GMF-KO and GMF-KO+Adv-LacZ mice.


Asunto(s)
Encefalomielitis Autoinmune Experimental/genética , Factor de Maduración de la Glia/genética , Factor de Maduración de la Glia/metabolismo , Animales , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Ensayo de Inmunoadsorción Enzimática , Glicoproteínas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos/inmunología , Médula Espinal/patología
5.
Brain Res ; 1208: 192-203, 2008 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-18395194

RESUMEN

Glia maturation factor (GMF), discovered and characterized in our laboratory, is a highly conserved protein primarily localized in mammalian central nervous system. Previously we demonstrated that GMF is required in the induced production of proinflammatory cytokines and chemokines in brain cells. We now report that ventricular infusion of human amyloid beta peptide1-42 (Abeta1-42) in mouse brain caused glial activation and large increases in the levels of GMF as well as induction of inflammatory cytokine/chemokine known for launching the neuro inflammatory cascade in Alzheimer's disease (AD). To test the hypothesis that GMF is involved in the pathogenesis of AD, we infused Abeta1-42 in the brain of GMF-deficient (GMF-KO) mice, recently prepared in our laboratory. GMF-deficient mice showed reduced glial activation and significantly suppressed proinflammatory cytokine/chemokine production following Abeta infusion compared to wild type (Wt) mice. The decrease in glial activation in the GMF-KO mice is also associated with significant reduction in Abeta induced loss of pre-synaptic marker, synaptophysin, and post-synaptic density protein-95 (PSD 95). We also examined the potential relationship between GMF or lack of it with learning and memory using the T-maze, Y-maze, and water maze, hippocampal-dependent spatial memory tasks. Our results show that memory retention was improved in GMF-KO mice compared to Wt controls following Abeta infusion. Diminution of these Abeta1-42 effects in primary cultures of GMF-KO astrocyte and microglia were reversed by reconstituted expression of GMF. Taken together, our results indicate a novel mediatory role of GMF in the neuro-inflammatory pathway of Abeta and its pro-inflammatory functions.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Citocinas/metabolismo , Factor de Maduración de la Glia/farmacología , Neuroglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Análisis de Varianza , Animales , Animales Recién Nacidos , Conducta Animal/efectos de los fármacos , Encéfalo/citología , Muerte Celular/efectos de los fármacos , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática/métodos , Factor de Maduración de la Glia/deficiencia , Humanos , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Noqueados , Neuroglía/fisiología , Neuronas/patología , Factores de Tiempo
6.
Neurochem Res ; 33(1): 22-6, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17577665

RESUMEN

PURPOSE: Fenretinide, 4-(N-hydroxyphenyl) retinamide, (4-HPR) is a well tolerated analog of alltrans retinoic acid. The gangliosideGM3, is a non-specific inhibitor of EGF receptor autophosphorylation (EGFR-phos). Both compounds were found preferentially cytotoxic to malignant and proliferating cells when compared to non-proliferating normal brain cells. Some of the small molecule inhibitors of EGFR-phos are also known to inhibit growth of brain tumors at relatively non-toxic doses. The purpose of this investigation was to evaluate if 4-HPR and inhibitors of EGFR-phos could be used together in the treatment of brain tumors. METHODS: The 9L rat gliosarcoma cells were treated in vitro with 4-HPR either alone or in combination with the non-specific or specific inhibitors of EGFR-phos, GM3 or AG-1478, respectively. The relative viability of the control and treated cells was determined using 3-(4,5-imethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. The experimental data were analyzed for statistical significance. RESULTS: In contrast to the expected additive/synergistic effect on cell growth inhibition, the sub-toxic and toxic concentrations of 4-HPR protected GM3 treated cells. The viable cells were 3.86 times higher following GM3 plus 4-HPR treatments compared to GM3 treatment alone. Additionally, a specific inhibitor of EGFR-phos signaling, AG-1478 caused a concentration dependent protection of cells from the toxicity of 4-HPR. Our results show counteracting cytotoxic responses of 4-HPR and EGFR-phos inhibitors when used together in 9L rat gliosarcoma cells.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Fenretinida/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular Tumoral , Receptores ErbB/metabolismo , Ratas
7.
Brain Res ; 1190: 206-14, 2008 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-18054898

RESUMEN

In the present study we report that a replication-defective adenovirus construct of GMF cDNA (GMF-V) induced overexpression of GMF protein in neuroblastoma (N18) cells caused cytotoxicity and loss of cell viability. A significant increase in activation of GSK-3beta occurred after infection with GMF-V when compared with mock and lacZ controls. Overexpression of GMF also increased caspase-3 activity, an early marker of apoptosis. Depletion of GMF gene by introducing GMF-specific siRNA (GsiRNA) completely blocked both activation of GSK-3beta and caspase-3 activation whereas a control scrambled siRNA (CsiRNA) had no effect. A cell-permeable peptide inhibitor of GSK-3beta, and lithium completely prevented GMF-dependent activation of caspase-3. These results demonstrate that GSK-3 mediates activation of the death domain caspase by GMF overexpression. We also show that the phosphorylation of GSK-3-dependent site of Tau was a consequence of GMF-overexpression in N18 cells. Taken together our results imply that GMF is involved in the signaling leading to the activation of GSK-3beta and caspase-3 in N18 cells and strongly suggest its involvement in neurodegeneration since GSK-3beta is known to hyperphosphorylate tau which is associated with the neurotoxicity of neurofibrillary tangles in Alzheimer's disease.


Asunto(s)
Apoptosis/fisiología , Caspasa 3/metabolismo , Factor de Maduración de la Glia/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Neuroblastoma/metabolismo , Animales , Línea Celular Tumoral , Regulación de la Expresión Génica/fisiología , Silenciador del Gen/fisiología , Glucógeno Sintasa Quinasa 3 beta , Ratones , ARN Interferente Pequeño/metabolismo , Transfección
8.
Neurosci Res ; 58(2): 156-63, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17383764

RESUMEN

In this study we show the effect of anti-sulfatide (RmAb) antibodies and inflammatory cytokines, TNF-alpha and IFN-gamma in inducing myelin basic protein (MBP) degradation in myelin isolated from control wild type (WT) and glia maturation factor (GMF)-deficient (GMF-KO) mice. GMF was not detected in isolated myelin from WT and GMF-KO mice although it is present in brains of WT mice. Our results show that calcium-dependent neutral protease activity caused significantly elevated degradation of 18.5 and/or 17.5kDa isoforms of MBP in WT myelin treated with RmAb or IFN-gamma. In contrast, MBP degradation in isolated myelin from GMF-KO mice remained unaffected following treatment with RmAb, IFN-gamma, or GM-CSF. Neither the 14kDa isoform of MBP nor proteolipid protein (PLP) showed an elevated degradation compared to controls. A virtual absence of GM-CSF, TNF-alpha and IFN-gamma in GMF-KO brain compared to WT was also evident when the animals were challenged with MOG 35-55. Additionally, the myelin from GMF-KO mice showed difference in distribution of myelin oligodendrocyte glycoprotein (MOG) and beta-tubulin in a sucrose density gradient myelin-axolemmal fractions compared to WT. Taken together, our data suggests a role for GMF in the biochemical organization of myelin and thereby its effect on MBP degradation induced by RmAb and IFN-gamma.


Asunto(s)
Anticuerpos/farmacología , Factor de Maduración de la Glia/deficiencia , Interferón gamma/farmacología , Proteína Básica de Mielina/metabolismo , Vaina de Mielina/efectos de los fármacos , Sulfoglicoesfingolípidos/inmunología , Animales , Calcio/metabolismo , Quelantes/farmacología , Modelos Animales de Enfermedad , Interacciones Farmacológicas , Ácido Egtácico/farmacología , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Masculino , Ratones , Ratones Noqueados , Proteínas de la Mielina , Glicoproteína Asociada a Mielina , Glicoproteína Mielina-Oligodendrócito , Neuritis Autoinmune Experimental/inducido químicamente , Neuritis Autoinmune Experimental/metabolismo
9.
Brain Res ; 1144: 239-47, 2007 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-17316572

RESUMEN

Pro-inflammatory cytokines/chemokines are implemented in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model with clinical and pathological similarities to multiple sclerosis. We have previously shown that over-expression of glia maturation factor (GMF) in glial cells cause excessive production and secretion of pro-inflammatory cytokines/chemokines sufficient to destroy the myelin-forming oligodendroglial cell in vitro. In this present investigation, we evaluate the expression of pro-inflammatory mediators in the central nervous system (CNS) of GMF+/+ (wild type) mice and GMF-/- (GMF-knockout) mice at the peak of EAE induced by immunization with MOG 35-55 peptide. GMF+/+ (Wt) mice developed severe EAE with a maximal mean clinical score of 3.6+/-0.5 by day 16 post-immunization, whereas GMF-KO mice showed significantly delayed EAE with an average onset on day 26 pi with reduced mean clinical score of 1.3+/-0.3. Three of fifteen Wt mice as compared to none of GMF-KO mice died of EAE. Encephalitogenic cells from Wt mice transferred to recipient GMF-KO mice caused very mild and with low incidence of EAE. We determined the differences in the expression of cytokines, IFN-gamma, TNF-alpha, IL-1 beta, IL-6, IL-4, IL-10, and chemokines, MIP-1, MIP-2, IP-10, MCP-1, GM-CSF mRNA by quantitative real-time RT-PCR in brain and spinal cord. Our results demonstrate significantly low levels of pro-inflammatory cytokines/chemokines in the CNS of GMF-KO mice and increased expression in Wt mice with EAE. Our data suggest that GMF play a critical role in CNS inflammation.


Asunto(s)
Sistema Nervioso Central/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Regulación de la Expresión Génica/fisiología , Factor de Maduración de la Glia/deficiencia , Análisis de Varianza , Animales , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/fisiopatología , Ensayo de Inmunoadsorción Enzimática/métodos , Regulación de la Expresión Génica/genética , Glicoproteínas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos , ARN Mensajero/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
10.
J Neurochem ; 101(2): 364-76, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17250654

RESUMEN

The glia maturation factor (GMF), which was discovered in our laboratory, is a highly conserved protein predominantly localized in astrocytes. GMF is an intracellular regulator of stress-related signal transduction. We now report that the overexpression of GMF in astrocytes leads to the destruction of primary oligodendrocytes by interactions between highly purified cultures of astrocytes, microglia, and oligodendrocytes. We infected astrocytes with a replication-defective adenovirus carrying the GMF cDNA. The overexpression of GMF caused the activation of p38 MAP kinase and transcription factor NF-kappaB, as well as the induction of granulocyte-macrophage colony-stimulating factor (GM-CSF) mRNA and protein in astrocytes. Small interfering RNA-mediated GMF knockdown completely blocked the GMF-dependent activation of p38 mitogen-activated protein kinase (MAPK), NF-kappaB, and enhanced expression of GM-CSF by astrocytes. Inhibition of p38 MAPK or NF-kappaB by specific inhibitors prevented GM-CSF production. The cell-free conditioned medium from overexpressing GMF astrocytes contained 320 +/- 33 pg/mL of GM-CSF, which was responsible for enhanced production and secretion of TNF-alpha, IL-1beta, IL-6, and IP-10 by microglia. Presence of these inflammatory cytokines in the conditioned medium from microglia efficiently destroyed oligodendrocytes in culture. These results suggest that GMF-induced production of GM-CSF in astrocytes is depending on p38 MAPK and NF-kappaB activation. The GM-CSF-dependent expression and secretion of inflammatory cytokine/chemokine, TNF-alpha, IL-1beta, IL-6, and IP-10, is cytotoxic to oligodendrocytes, the myelin-forming cells in the central nervous system, and as well as neurons. Our results suggest a novel pathway of GMF-initiated cytotoxicity of brain cells, and implicate its involvement in inflammatory diseases such as multiple sclerosis.


Asunto(s)
Astrocitos/metabolismo , Citocinas/metabolismo , Encefalitis/metabolismo , Factor de Maduración de la Glia/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Microglía/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/inmunología , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/fisiopatología , Muerte Celular/inmunología , Células Cultivadas , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Citocinas/toxicidad , Encefalitis/inmunología , Encefalitis/fisiopatología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/inmunología , Factor de Maduración de la Glia/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Ratones , Microglía/inmunología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Degeneración Nerviosa/inmunología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Oligodendroglía/inmunología , Oligodendroglía/metabolismo , ARN Interferente Pequeño , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Neurochem Res ; 32(1): 115-24, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17160503

RESUMEN

Cis-parinaric acid (c-PNA), a natural four conjugated polyunsaturated fatty acid, increases free radical production and it is preferentially cytotoxic to malignant glial cells compared to normal astrocytes in-vitro. In order to explain the increased cytotoxicity of c-PNA in malignant glial cells, we compared the effects of c-PNA on the oxidative stress-dependent signal transducing events in 36B10 cells, a malignant rat astrocytoma cell line, and in fetal rat astrocytes. Our results show that c-PNA treatment in 36B10 cells caused a persistent activation of c-Jun N-terminal protein kinase (JNK) at RNA and protein levels. Specific inhibitors of the kinase significantly reversed the cytotoxicity of c-PNA. Additionally, c-PNA caused the phosphorylated inactivation of forkhead transcription factor-3a (FKHR-L1, FOXO3a) and drastically decreased the activity of mitochondrial superoxide dismutase (Mn-SOD) that protects cells from oxidative stress. On the other hand, identical c-PNA treatments in normal astrocytes increased the dephosphorylated activation of FKHR-L1, maintained activity of Mn-SOD and failed to phosphorylate JNK. Taken together, the results imply that a selective activation of JNK and the opposite regulation of FKHR-L1 and Mn-SOD contribute to the differential cytotoxicity of c-PNA in malignant and normal glial cells.


Asunto(s)
Astrocitos/efectos de los fármacos , Ácidos Grasos Insaturados/toxicidad , Factores de Transcripción Forkhead/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Animales , Astrocitos/metabolismo , Astrocitoma/tratamiento farmacológico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Curcumina/farmacología , Flavonoides/farmacología , Proteína Forkhead Box O3 , Radicales Libres/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Fosforilación , Ratas , Ratas Endogámicas F344 , Superóxido Dismutasa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos
12.
Neurochem Res ; 32(1): 39-47, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17151915

RESUMEN

Glia maturation factor (GMF), a highly conserved brain-specific protein, isolated, sequenced and cloned in our laboratory. Overexpression of GMF in astrocytes induces the production and secretion of granulocyte-macrophage-colony stimulating factor (GM-CSF), and subsequent immune activation of microglia, expression of several proinflammatory genes including major histocompatibility complex proteins, IL-1beta, and MIP-1beta, all associated with the development of experimental autoimmune encephalomyelitis (EAE), the animal model for multiple sclerosis. Based on GMF's ability to activate microglia and induce well-established proinflammatory mediators, including GM-CSF, we hypothesize that GMF is involved in the pathogenesis of inflammatory disease EAE. In this present investigation, using GMF-deficient mice, we study the role of GMF and how the lack of GMF affects the EAE disease. Our results show a significant decrease in incidence, delay in onset, and reduced severity of EAE in GMF-deficient mice, and support the hypothesis that GMF plays a major role in the pathogenesis of disease.


Asunto(s)
Encefalomielitis Autoinmune Experimental/fisiopatología , Factor de Maduración de la Glia/deficiencia , Animales , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Ratones , Ratones Noqueados , Proteínas de la Mielina , Glicoproteína Asociada a Mielina/inmunología , Glicoproteína Mielina-Oligodendrócito , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Fragmentos de Péptidos/farmacología , Factor de Necrosis Tumoral alfa/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...