Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Virology ; 573: 131-140, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35779335

RESUMEN

Japanese Encephalitis Virus (JEV), a member virus of Flaviviridae family causes Japanese encephalitis (JE). JE is a mosquito-borne disease, spread mainly by Culex spp. During JE, dysregulated inflammatory responses play a central role in neuronal death and damage leading to Neuroinflammation. In this study, we show that JEV infection in human microglial cells (CHME3) reduces the cellular miR-590-3p levels. miR-590-3p could directly target the expression levels of USP42 (Ubiquitin Specific Peptidase 42) resulting in increased cellular levels of USP42 upon JEV infection. Our results suggest that USP42 stabilizes cellular TRIM21 via deubiquitinating them. We also established through various in vitro and in vivo experiments that increased USP42 can maintain a higher cellular level of both TRIM21 as well as OAS1. This study also suggests that TRIM21, independently of its RING domain, can increase USP42 level in a positive feedback loop and induces the cellular OAS1 levels in human microglial cells.


Asunto(s)
2',5'-Oligoadenilato Sintetasa , Encefalitis Japonesa , Ribonucleoproteínas , Tioléster Hidrolasas , 2',5'-Oligoadenilato Sintetasa/metabolismo , Animales , Virus de la Encefalitis Japonesa (Especie) , Humanos , MicroARNs/metabolismo , Microglía/metabolismo , Ribonucleoproteínas/metabolismo , Tioléster Hidrolasas/metabolismo
3.
Sci Signal ; 10(466): eaaf5185, 2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28196914

RESUMEN

Effective recognition of viral components and the subsequent stimulation of the production of type I interferons (IFNs) is crucial for the induction of host antiviral immunity. The failure of the host to efficiently produce type I IFNs in response to infection by the Japanese encephalitis virus (JEV) is linked with an increased probability for the disease to become lethal. JEV is a neurotropic virus of the Flaviviridae family that causes encephalitis in humans. JEV infection is regulated by several host factors, including microRNAs, which are conserved noncoding RNAs that participate in various physiological and pathological processes. We showed that the JEV-induced expression of miR-301a led to inhibition of the production of type I IFN by reducing the abundances of the transcription factor IFN regulatory factor 1 (IRF1) and the signaling protein suppressor of cytokine signaling 5 (SOCS5). Mechanistically, induction of miR-301a expression during JEV infection required the transcription factor nuclear factor κB. In mouse neurons, neutralization of miR-301a restored the host innate immune response by enabling IFN-ß production, thereby restricting viral propagation. Inhibition of miR-301a in mouse brain rescued the production of IRF1 and SOCS5, increased the generation of IFN-ß, and reduced the extent of JEV replication, thus improving mouse survival. Thus, our study suggests that the JEV-induced expression of miR-301a assists viral pathogenesis by suppressing IFN production, which might be targeted by antiviral therapies.


Asunto(s)
Inmunidad Innata/genética , Factor 1 Regulador del Interferón/genética , MicroARNs/genética , Neuronas/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Virus de la Encefalitis Japonesa (Especie)/fisiología , Femenino , Perfilación de la Expresión Génica , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Factor 1 Regulador del Interferón/metabolismo , Masculino , Ratones Endogámicos BALB C , Neuronas/virología , Transducción de Señal/genética , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
4.
J Neuroinflammation ; 12: 30, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25889446

RESUMEN

BACKGROUND: Japanese encephalitis virus (JEV) is the causative agent of Japanese encephalitis which is more prevalent in South and Southeast Asia. JEV is a neurotropic virus which infiltrates into the brain through vascular endothelial cells. JEV infects neurons and microglial cells which causes neuronal damage and inflammation. However, JEV also evades the cellular immune response to survive in host cells. Viruses are known to modulate the expression of microRNAs, which in turn modulate cellular immune response by targeting expression of antiviral genes. The aim of this study is to understand the anti-inflammatory role of miR-146a during JEV infection, which facilitates immune evasion. METHODS: Human brain microglial cells (CHME3) were infected by JEV: JaOArS982 and P20778 strain, and expression of miR-146a were analyzed. Overexpression and knockdown studies of miR-146a were done to see the effect on NF-κB pathway and antiviral Jak-STAT pathway. Regulatory role of miR-146a on expression of interferon-stimulated genes was determined by real-time PCR and luciferase assays. RESULTS: JEV infection elevated the expression of miR-146a in JaOArS982 strain which caused downregulation of TRAF6, IRAK1, IRAK2, and STAT1 genes. Exogenous overexpression of miR-146a led to suppression of NF-κB activation and abrogation of Jak-STAT pathway upon JEV infection which led to downregulation of interferon-stimulated genes (IFIT-1 and IFIT-2) and facilitated viral replication. JEV infection initially upregulated cytokine production and activated STAT1 activity but STAT1 levels reduced at later time point, which led to the downregulation of interferon-stimulated genes. CONCLUSION: Upregulation of miR-146a by JEV JaOArS982 strain leads to suppression of NF-κB activity and disruption of antiviral Jak-STAT signaling which helps the virus to evade the cellular immune response. This effect of JEV infection on miR-146a expression was found to be strain specific.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/inmunología , Regulación Viral de la Expresión Génica/fisiología , MicroARNs/metabolismo , Microglía/inmunología , Microglía/virología , Línea Celular Transformada , Citocinas/metabolismo , Humanos , Inmunidad Celular , Quinasas Asociadas a Receptores de Interleucina-1/genética , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , MicroARNs/genética , FN-kappa B/metabolismo , ARN Mensajero/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Factores de Tiempo
5.
J Neuroinflammation ; 11: 24, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24485101

RESUMEN

BACKGROUND: Japanese encephalitis virus (JEV) infection leads to Japanese encephalitis (JE) in humans. JEV is transmitted through mosquitoes and maintained in a zoonotic cycle. This cycle involves pigs as the major reservoir, water birds as carriers and mosquitoes as vectors. JEV invasion into the central nervous system (CNS) may occur via antipodal transport of virions or through the vascular endothelial cells. Microglial cells get activated in response to pathogenic insults. JEV infection induces the innate immune response and triggers the production of type I interferons. The signaling pathway of type I interferon production is regulated by a number of molecules. TRIM proteins are known to regulate the expression of interferons; however, the involvement of TRIM genes and their underlying mechanism during JEV infection are not known. METHODS: Human microglial cells (CHME3) were infected with JEV to understand the role of TRIM21 in JEV infection and its effect on type I interferon (IFN-ß) production. Cells were infected in presence and absence of exogenous TRIM21 as well as after knocking down the TRIM21 mRNA. Levels of activated IRF3 expression were measured through Western blot analyses of anti-p-IRF3 antibody, and IFN-ß production was measured by using IFN-ß real-time PCR and luciferase activity analyses. RESULTS: JEV infection increased expression of TRIM21 in CHME3 cells. JEV induced an innate immune response by increasing production of IFN-ß via IRF3 activation and phosphorylation. Overexpression of TRIM21 resulted in downregulation of p-IRF3 and IFN-ß, while silencing led to increased production of p-IRF3 and IFN-ß in JEV-infected CHME3 cells. CONCLUSION: This report demonstrates TRIM21 as a negative regulator of interferon-ß (IFN-ß) production mediated by IRF-3 during JEV infection in human microglial cells.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/fisiología , Regulación Viral de la Expresión Génica/fisiología , Interferón Tipo I/metabolismo , Microglía/metabolismo , Ribonucleoproteínas/metabolismo , Transducción de Señal/fisiología , Línea Celular Transformada , Humanos , Factor 3 Regulador del Interferón/metabolismo , Microglía/virología , Fosforilación , ARN Mensajero , Factores de Tiempo , Transfección , Ensayo de Placa Viral
6.
Neurotoxicology ; 41: 143-53, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24530660

RESUMEN

Lead is one of the major pollutants of environment and is highly toxic to the functioning of central nervous system (CNS). The chronic exposure of this heavy metal is debilitating to the functional behavior of an organism. Studies have shown that acute exposure to Pb can lead to glial activation and secretion of cyto-chemokines in both in vitro and in vivo models. However, the cellular source of secretion of these cyto-chemokines remains to be identified. Microglia are monocytes of the brain, and are primary source of cytokine secretion in the CNS. We hypothesized that microglia exposed to Pb can secrete cyto-chemokines, thereby resulting in subsequent neuronal death. Our studies show that stimulation of BV-2 mouse microglia with 10µÐœ dose of Pb resulted in up-regulation of extracellular signal-regulated kinase (ERK) and protein kinase B (Akt) pathways, along with activation of an important transcription factor, nuclear factor-κB (NF-κB). Further, we found that the levels of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and monocyte chemoattractant protein-1 (MCP-1), and cyclooxygenase-2 (COX-2) pro-inflammatory enzyme were increased in response to Pb exposure. Furthermore, treatment with conditioned media from Pb treated BV-2 cells lead to neuronal death in neuroblastoma cells, which potentially involved the activation of caspase-3 enzyme. In all, the current study brings forth critical involvement of microglial activation in mediating the neurotoxicity associated with lead exposure.


Asunto(s)
Microglía/efectos de los fármacos , Neuronas/fisiología , Compuestos Organometálicos/farmacología , Análisis de Varianza , Animales , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Transformada , Supervivencia Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , MAP Quinasa Quinasa Quinasa 5/metabolismo , Ratones , Peso Molecular , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
7.
PLoS Negl Trop Dis ; 7(1): e2005, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23350007

RESUMEN

BACKGROUND: Japanese encephalitis virus (JEV) is a major cause of viral encephalitis in South and South-East Asia. Lack of antivirals and non-availability of affordable vaccines in these endemic areas are a major setback in combating JEV and other closely related viruses such as West Nile virus and dengue virus. Protein secondary structure mimetics are excellent candidates for inhibiting the protein-protein interactions and therefore serve as an attractive tool in drug development. We synthesized derivatives containing the backbone of naturally occurring lupin alkaloid, sparteine, which act as protein secondary structure mimetics and show that these compounds exhibit antiviral properties. METHODOLOGY/PRINCIPAL FINDINGS: In this study we have identified 3,7-diazabicyclo[3.3.1]nonane, commonly called bispidine, as a privileged scaffold to synthesize effective antiviral agents. We have synthesized derivatives of bispidine conjugated with amino acids and found that hydrophobic amino acid residues showed antiviral properties against JEV. We identified a tryptophan derivative, Bisp-W, which at 5 µM concentration inhibited JEV infection in neuroblastoma cells by more than 100-fold. Viral inhibition was at a stage post-entry and prior to viral protein translation possibly at viral RNA replication. We show that similar concentration of Bisp-W was capable of inhibiting viral infection of two other encephalitic viruses namely, West Nile virus and Chandipura virus. CONCLUSIONS/SIGNIFICANCE: We have demonstrated that the amino-acid conjugates of 3,7-diazabicyclo[3.3.1]nonane can serve as a molecular scaffold for development of potent antivirals against encephalitic viruses. Our findings will provide a novel platform to develop effective inhibitors of JEV and perhaps other RNA viruses causing encephalitis.


Asunto(s)
Aminoácidos/farmacología , Antivirales/farmacología , Materiales Biomiméticos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Virus de la Encefalitis Japonesa (Especie)/fisiología , Replicación Viral/efectos de los fármacos , Aminoácidos/química , Animales , Antivirales/química , Materiales Biomiméticos/química , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Línea Celular , Hepatocitos/virología , Humanos , Pruebas de Sensibilidad Microbiana , Neuronas/virología , Vesiculovirus/efectos de los fármacos , Vesiculovirus/fisiología , Virus del Nilo Occidental/efectos de los fármacos , Virus del Nilo Occidental/fisiología
8.
PLoS One ; 7(4): e35427, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22514742

RESUMEN

BACKGROUND: Japanese encephalitis (JE), the most common form of viral encephalitis occurs periodically in endemic areas leading to high mortality and neurological deficits in survivors. It is caused by a flavivirus, Japanese encephalitis virus (JEV), which is transmitted to humans through mosquitoes. No effective cure exists for reducing mortality and morbidity caused by JEV infection, which is primarily due to excessive inflammatory response. Fenofibrate, a peroxisome proliferator-activated receptor-α (PPARα) agonist is known to resolve inflammation by repressing nuclear factor-κB (NF-κB) and enhancing transcription of anti-oxidant and anti-inflammatory genes. In addition, fenofibrate also up-regulates a class of proteins, cytochrome P4504Fs (Cyp4fs), which are involved in detoxification of the potent pro-inflammatory eicosanoid, leukotriene B(4) (LTB(4)) to 20-hydroxy LTB(4). METHODOLOGY/PRINCIPAL FINDINGS: The neuroprotective effect of fenofibrate was examined using in vitro (BV-2 microglial cell line) and in vivo (BALB/c mice) models of JEV infection. Mice were treated with fenofibrate for 2 or 4 days prior to JEV exposure. Pretreatment with fenofibrate for 4 but not 2 days reduced mortality by 80% and brain LTB(4) levels decreased concomitantly with the induction of Cyp4f15 and 4f18, which catalyze detoxification of LTB(4) through hydroxylation. Expression of cytokines and chemokine decreased significantly as did microglial activation and replication of the JEV virus. CONCLUSIONS/SIGNIFICANCE: Fenofibrate confers neuroprotection against Japanese encephalitis, in vivo, in mouse model of JEV infection. Thus, fenofibrate, a PPARα agonist that is commonly used as a hypolipidemic drug could potentially be used for prophylaxis during JE epidemics to reduce mortality and morbidity.


Asunto(s)
Encefalitis Japonesa/tratamiento farmacológico , Encefalitis Japonesa/metabolismo , Fenofibrato/uso terapéutico , Animales , Quimiocinas/metabolismo , Citocinas/metabolismo , Eicosanoides/metabolismo , Encefalitis Japonesa/virología , Leucotrieno B4/metabolismo , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , PPAR alfa/agonistas , Receptores Activados del Proliferador del Peroxisoma/metabolismo
9.
J Neuroinflammation ; 9: 57, 2012 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-22429472

RESUMEN

BACKGROUND: Neuroinflammation occurs as a result of microglial activation in response to invading micro-organisms or other inflammatory stimuli within the central nervous system. According to our earlier findings, Krüppel-like factor 4 (Klf4), a zinc finger transcription factor, is involved in microglial activation and subsequent release of proinflammatory cytokines, tumor necrosis factor alpha, macrophage chemoattractant protein-1 and interleukin-6 as well as proinflammatory enzymes, inducible nitric oxide synthase and cyclooxygenase-2 in lipopolysaccharide-treated microglial cells. Our current study focuses on finding the molecular mechanism of the anti-inflammatory activities of honokiol in lipopolysaccharide-treated microglia with emphasis on the regulation of Klf4. METHODS: For in vitro studies, mouse microglial BV-2 cell lines as well as primary microglia were treated with 500 ng/mL lipopolysaccharide as well as 1 µM and 10 µM of honokiol. We cloned full-length Klf4 cDNA in pcDNA3.1 expression vector and transfected BV-2 cells with this construct using lipofectamine for overexpression studies. For in vivo studies, brain tissues were isolated from BALB/c mice treated with 5 mg/kg body weight of lipopolysaccharide either with or without 2.5 or 5 mg/kg body weight of honokiol. Expression of Klf4, cyclooxygenase-2, inducible nitric oxide synthase and phospho-nuclear factor-kappa B was measured using immunoblotting. We also measured the levels of cytokines, reactive oxygen species and nitric oxide in different conditions. RESULTS: Our findings suggest that honokiol can substantially downregulate the production of proinflammatory cytokines and inflammatory enzymes in lipopolysaccharide-stimulated microglia. In addition, honokiol downregulates lipopolysaccharide-induced upregulation of both Klf4 and phospho-nuclear factor-kappa B in these cells. We also found that overexpression of Klf4 in BV-2 cells suppresses the anti-inflammatory action of honokiol. CONCLUSIONS: Honokiol potentially reduces inflammation in activated microglia in a Klf4-dependent manner.


Asunto(s)
Inflamación/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Microglía/efectos de los fármacos , Animales , Línea Celular Transformada , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inflamación/inducido químicamente , Factor 4 Similar a Kruppel , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Polisacáridos/efectos adversos , Polisacáridos/farmacología , Especies Reactivas de Oxígeno/metabolismo
10.
PLoS One ; 7(2): e32270, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22393394

RESUMEN

BACKGROUND: Japanese Encephalitis virus (JEV) is a common cause of acute and epidemic viral encephalitis. JEV infection is associated with microglial activation resulting in the production of pro-inflammatory cytokines including Interleukin-1 ß (IL-1ß) and Interleukin-18 (IL-18). The Pattern Recognition Receptors (PRRs) and the underlying mechanism by which microglia identify the viral particle leading to the production of these cytokines is unknown. METHODOLOGY/PRINCIPAL FINDINGS: For our studies, we have used murine model of JEV infection as well as BV-2 mouse microglia cell line. In this study, we have identified a signalling pathway which leads to the activation of caspase-1 as the key enzyme responsible for the maturation of both IL-1ß and IL-18 in NACHT, LRR and PYD domains-containing protein-3 (NLRP3) dependent manner. Depletion of NLRP3 results in the reduction of caspase-1 activity and subsequent production of these cytokines. CONCLUSION/SIGNIFICANCE: Our results identify a mechanism mediated by Reactive Oxygen Species (ROS) production and potassium efflux as the two danger signals that link JEV infection to caspase-1 activation resulting in subsequent IL-1ß and IL-18 maturation.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Virus de la Encefalitis Japonesa (Especie)/metabolismo , Encefalitis Japonesa/genética , Encefalitis Japonesa/virología , Animales , Caspasa 1/metabolismo , Línea Celular , Citocinas/metabolismo , Inflamación , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos BALB C , Microglía/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Estructura Terciaria de Proteína , Especies Reactivas de Oxígeno , Transducción de Señal
11.
PLoS One ; 6(3): e17225, 2011 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-21390230

RESUMEN

BACKGROUND: Japanese encephalitis virus (JEV) induces neuroinflammation with typical features of viral encephalitis, including inflammatory cell infiltration, activation of microglia, and neuronal degeneration. The detrimental effects of inflammation on neurogenesis have been reported in various models of acute and chronic inflammation. We investigated whether JEV-induced inflammation has similar adverse effects on neurogenesis and whether those effects can be reversed using an anti-inflammatory compound minocycline. METHODOLOGY/PRINCIPAL FINDINGS: Here, using in vitro studies and mouse models, we observed that an acute inflammatory milieu is created in the subventricular neurogenic niche following Japanese encephalitis (JE) and a resultant impairment in neurogenesis occurs, which can be reversed with minocycline treatment. Immunohistological studies showed that proliferating cells were replenished and the population of migrating neuroblasts was restored in the niche following minocycline treatment. In vitro, we checked for the efficacy of minocycline as an anti-inflammatory compound and cytokine bead array showed that production of cyto/chemokines decreased in JEV-activated BV2 cells. Furthermore, mouse neurospheres grown in the conditioned media from JEV-activated microglia exhibit arrest in both proliferation and differentiation of the spheres compared to conditioned media from control microglia. These effects were completely reversed when conditioned media from JEV-activated and minocycline treated microglia was used. CONCLUSION/SIGNIFICANCE: This study provides conclusive evidence that JEV-activated microglia and the resultant inflammatory molecules are anti-proliferative and anti-neurogenic for NSPCs growth and development, and therefore contribute to the viral neuropathogenesis. The role of minocycline in restoring neurogenesis may implicate enhanced neuronal repair and attenuation of the neuropsychiatric sequelae in JE survivors.


Asunto(s)
Encefalitis Japonesa/complicaciones , Encefalitis Japonesa/patología , Inflamación/complicaciones , Inflamación/patología , Neurogénesis , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/virología , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quimiocinas/biosíntesis , Modelos Animales de Enfermedad , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Virus de la Encefalitis Japonesa (Especie)/fisiología , Encefalitis Japonesa/virología , Mediadores de Inflamación/metabolismo , Ratones , Microglía/efectos de los fármacos , Microglía/patología , Minociclina/farmacología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/patología , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/patología , Solubilidad/efectos de los fármacos , Virión/efectos de los fármacos , Virión/metabolismo
12.
J Neuroimmune Pharmacol ; 5(4): 553-65, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20635157

RESUMEN

Japanese encephalitis (JE) is caused by a neurotropic flavivirus that causes CNS damage that leads to death in acute cases or permanent neuropsychiatric sequel in survivors. The course of infection of this virus is not well defined though it is clear that it evades the host's innate immune response in the periphery. The current study was designed to investigate the time-dependent changes in the spleen and lymph node, apart from the CNS that are infected by the Japanese encephalitis virus (JEV). Our previous studies have led to the identification of minocycline, a semi-synthetic antibiotic, as a protective drug in JE. In this study we have also investigated the role of minocycline on the peripheral organs that are infected by JEV. Levels of IL-12 and MCP-1 in the organs were estimated by cytometric bead array, and immunohistochemical studies were performed on cryosections of tissue to detect CD3- or CD11b-positive cells as well as JEV antigen. We found that the levels of T cell-activating cytokine IL-12 and MCP-1 levels were significantly elevated in JEV-infected tissue samples in a time-dependent manner. Corresponding to this increase was the increase in the number of CD3- and CD11b-positive cells in the tissues of infected animals. Minocycline treatment abrogated these changes. Minocycline treatment also resulted in the gradual decrease in the number of CD11b (but not CD3) positive cells in the lymph node and spleen, even though the virus persisted in these organs. We also observed structural changes in the spleen following minocycline treatment.


Asunto(s)
Antibacterianos/farmacología , Encefalitis Japonesa/tratamiento farmacológico , Encefalitis Japonesa/inmunología , Ganglios Linfáticos/efectos de los fármacos , Minociclina/farmacología , Bazo/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/virología , Quimiocina CCL2/biosíntesis , Modelos Animales de Enfermedad , Encefalitis Japonesa/virología , Inmunohistoquímica , Interleucina-12/biosíntesis , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/virología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Bazo/inmunología , Bazo/virología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
13.
PLoS One ; 5(4): e9984, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20376308

RESUMEN

BACKGROUND: Benzo[a]pyrene (B[a]P) belongs to a class of polycyclic aromatic hydrocarbons that serve as micropollutants in the environment. B[a]P has been reported as a probable carcinogen in humans. Exposure to B[a]P can take place by ingestion of contaminated (especially grilled, roasted or smoked) food or water, or inhalation of polluted air. There are reports available that also suggests neurotoxicity as a result of B[a]P exposure, but the exact mechanism of action is unknown. METHODOLOGY/PRINCIPAL FINDINGS: Using neuroblastoma cell line and primary cortical neuron culture, we demonstrated that B[a]P has no direct neurotoxic effect. We utilized both in vivo and in vitro systems to demonstrate that B[a]P causes microglial activation. Using microglial cell line and primary microglial culture, we showed for the first time that B[a]P administration results in elevation of reactive oxygen species within the microglia thereby causing depression of antioxidant protein levels; enhanced expression of inducible nitric oxide synthase, that results in increased production of NO from the cells. Synthesis and secretion of proinflammatory cytokines were also elevated within the microglia, possibly via the p38MAP kinase pathway. All these factors contributed to bystander death of neurons, in vitro. When administered to animals, B[a]P was found to cause microglial activation and astrogliosis in the brain with subsequent increase in proinflammatory cytokine levels. CONCLUSIONS/SIGNIFICANCE: Contrary to earlier published reports we found that B[a]P has no direct neurotoxic activity. However, it kills neurons in a bystander mechanism by activating the immune cells of the brain viz the microglia. For the first time, we have provided conclusive evidence regarding the mechanism by which the micropollutant B[a]P may actually cause damage to the central nervous system. In today's perspective, where rising pollution levels globally are a matter of grave concern, our study throws light on other health hazards that such pollutants may exert.


Asunto(s)
Benzo(a)pireno/farmacología , Carcinógenos/farmacología , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Benzo(a)pireno/toxicidad , Carcinógenos/toxicidad , Muerte Celular/efectos de los fármacos , Línea Celular , Células Cultivadas , Citocinas/biosíntesis , Citocinas/metabolismo , Contaminantes Ambientales/farmacología , Contaminantes Ambientales/toxicidad , Mediadores de Inflamación , Ratones , Microglía/metabolismo , Neuroblastoma/patología , Neuronas/citología , Óxido Nítrico/biosíntesis , Especies Reactivas de Oxígeno/metabolismo
14.
Immunobiology ; 215(11): 884-93, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20153075

RESUMEN

Japanese encephalitis virus (JEV) is a neurotropic flavivirus that is the causative agent of a major mosquito-borne encephalitis in the world. Evasion of peripheral immune system facilitates the entry of the virus into the central nervous system (CNS) where it causes extensive neuronal inflammatory damage that leads to death or severe neuropschychiatric sequel in survivors. It has been proposed that after entry into the body, the virus is carried into the CNS by peripheral immune cells that act as Trojan horses. In this study we investigate whether macrophages can be considered as such a Trojan horse. We also investigate the role of minocycline, a synthetic tetracycline, in such processes. Minocycline has been found to be broadly protective in neurological disease models featuring inflammation and cell death but there has been no report of it having any modulatory role in peripheral macrophage-mediated immune response against viral infection. Persistence of internalized virus within macrophages was visualized by immunofluorescent staining. Cytotoxicity assay revealed that there was no significant cell death after 24 h and 72 h infection with JEV. Proinflammatory cytokine levels were elevated in cells that were infected with JEV but it was abrogated following minocycline treatment. Reactive oxygen species level was also increased after JEV infection. Nitric oxide level was found to increase after 72 h post infection but remained unchanged after 24h. The cellular levels of signaling molecules such as PI3 kinase, phophoAkt and phospho p38MAP kinase were found to be altered after JEV infection and minocycline treatment. JEV infection also affected the VEGF-MMP pathway. Increased activity of MMP-9 was detected from JEV-infected macrophage culture supernatants after 72 h; minocycline treatment resulted in reduced activity. Thus it seems that minocycline dampens peripheral immune reactions by decreasing proinflammatory cytokine release from infected macrophages and the virus survives within macrophages long enough to be carried into the CNS, even though minocycline inhibits cell survival.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/fisiología , Encefalitis Japonesa/inmunología , Factores Inmunológicos/farmacología , Macrófagos/inmunología , Macrófagos/virología , Minociclina/farmacología , Animales , Células Cultivadas , Citocinas/metabolismo , Virus de la Encefalitis Japonesa (Especie)/patogenicidad , Femenino , Macrófagos/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Virulencia , Internalización del Virus
15.
Cell Biol Int ; 32(12): 1506-13, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18801452

RESUMEN

Astrocytes become activated in response to many CNS pathologies. The process of astrocyte activation remains rather enigmatic and results in so-called reactive gliosis, a reaction with specific structural and functional characteristics. Astrocytes play a vital role in regulating aspects of inflammation and in the homeostatic maintenance of the CNS. However, the responses of different human astroglial cell-lines in viral encephalitis mediated inflammation are not well documented. We have shown that Japanese encephalitis virus (JEV) infection causes morphological and functional changes in astrocytic cell-lines. We have demonstrated that besides reactive oxygen species (ROS) JEV infection differentially regulated the induction pattern of IL-6, IL-1 beta and IL-8. IP-10, MCP-1, MIG and RANTES secretions in different astroglial cell-lines. The expression of different proteins such as astrocyte-specific glial fibrillary acidic protein (GFAP), the glutamate aspartate transporter/essential amino acid transporter-1 (GLAST/EAAT-1), glutamate transporter-1/essential amino acid transporter-2 (GLT-1/EAAT-2), Ceruloplasmin and Thioredoxin (TRX) expression level also differ in different human astrocyte cell-lines following infection.


Asunto(s)
Astrocitos/inmunología , Encéfalo/inmunología , Encefalitis Japonesa/inmunología , Gliosis/inmunología , Estrés Oxidativo/inmunología , Animales , Animales Recién Nacidos , Astrocitos/virología , Astrocitoma/inmunología , Encéfalo/fisiopatología , Encéfalo/virología , Neoplasias Encefálicas/inmunología , Línea Celular Transformada , Línea Celular Tumoral , Ceruloplasmina/inmunología , Ceruloplasmina/metabolismo , Quimiocinas/inmunología , Quimiocinas/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Virus de la Encefalitis Japonesa (Especie)/inmunología , Encefalitis Japonesa/fisiopatología , Gliosis/fisiopatología , Gliosis/virología , Humanos , Ratones , Ratones Endogámicos BALB C , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Tiorredoxinas/inmunología , Tiorredoxinas/metabolismo , Proteínas de Transporte Vesicular de Glutamato/inmunología , Proteínas de Transporte Vesicular de Glutamato/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...