Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Autism ; 14(1): 43, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37968722

RESUMEN

BACKGROUND: Autism spectrum disorders (ASD) are predominantly neurodevelopmental and largely genetically determined. However, there are human data supporting the idea that fever can improve symptoms in some individuals, but those data are limited and there are almost no data to support this from animal models. We aimed to test the hypothesis that elevated body temperature would improve function in two animal models of ASD. METHODS: We used a 4 h whole-body hyperthermia (WBH) protocol and, separately, systemic inflammation induced by bacterial endotoxin (LPS) at 250 µg/kg, to dissociate temperature and inflammatory elements of fever in two ASD animal models: C58/J and Shank3B- mice. We used one- or two-way ANOVA and t-tests with normally distributed data and Kruskal-Wallis or Mann-Whitney with nonparametric data. Post hoc comparisons were made with a level of significance set at p < 0.05. For correlation analyses, data were adjusted by a linear regression model. RESULTS: Only LPS induced inflammatory signatures in the brain while only WBH produced fever-range hyperthermia. WBH reduced repetitive behaviours and improved social interaction in C58/J mice and significantly reduced compulsive grooming in Shank3B- mice. LPS significantly suppressed most activities over 5-48 h. LIMITATIONS: We show behavioural, cellular and molecular changes, but provide no specific mechanistic explanation for the observed behavioural improvements. CONCLUSIONS: The data are the first, to our knowledge, to demonstrate that elevated body temperature can improve behavioural signs in 2 distinct ASD models. Given the developmental nature of ASD, evidence that symptoms may be improved by environmental perturbations indicates possibilities for improving function in these individuals. Since experimental hyperthermia in patients would carry significant risks, it is now essential to pursue molecular mechanisms through which hyperthermia might bring about the observed benefits.


Asunto(s)
Trastorno del Espectro Autista , Hipertermia Inducida , Humanos , Ratones , Animales , Trastorno del Espectro Autista/terapia , Lipopolisacáridos/toxicidad , Temperatura , Modelos Animales de Enfermedad , Ratones Endogámicos , Encéfalo , Hipertermia Inducida/métodos
2.
Alzheimers Dement ; 17(10): 1735-1755, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34080771

RESUMEN

Neuroinflammation contributes to Alzheimer's disease (AD) progression. Secondary inflammatory insults trigger delirium and can accelerate cognitive decline. Individual cellular contributors to this vulnerability require elucidation. Using APP/PS1 mice and AD brain, we studied secondary inflammatory insults to investigate hypersensitive responses in microglia, astrocytes, neurons, and human brain tissue. The NLRP3 inflammasome was assembled surrounding amyloid beta, and microglia were primed, facilitating exaggerated interleukin-1ß (IL-1ß) responses to subsequent LPS stimulation. Astrocytes were primed to produce exaggerated chemokine responses to intrahippocampal IL-1ß. Systemic LPS triggered microglial IL-1ß, astrocytic chemokines, IL-6, and acute cognitive dysfunction, whereas IL-1ß disrupted hippocampal gamma rhythm, all selectively in APP/PS1 mice. Brains from AD patients with infection showed elevated IL-1ß and IL-6 levels. Therefore, amyloid leaves the brain vulnerable to secondary inflammation at microglial, astrocytic, neuronal, and cognitive levels, and infection amplifies neuroinflammatory cytokine synthesis in humans. Exacerbation of neuroinflammation to produce deleterious outcomes like delirium and accelerated disease progression merits careful investigation in humans.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Astrocitos/metabolismo , Inflamación/inmunología , Interleucina-1beta/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Amiloide/metabolismo , Animales , Encéfalo , Citocinas/metabolismo , Hipocampo , Humanos , Inflamasomas , Ratones , Ratones Transgénicos
3.
Front Cell Neurosci ; 14: 249, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32848629

RESUMEN

BACKGROUND: The immune system of human and mouse neonates is relatively immature. However, innate lymphoid cells (ILCs), commonly divided into the subsets ILC1, ILC2, and ILC3, are already present in the placenta and other fetal compartments and exhibit higher activity than what is seen in adulthood. Recent reports have suggested the potential role of ILCs, especially ILC2s, in spontaneous preterm labor, which is associated with brain damage and subsequent long-term neurodevelopmental deficits. Therefore, we hypothesized that ILCs, and especially ILC2s, play a role in preterm brain injury. METHODS: C57Bl/6J mice at postnatal day 6 were subjected to hypoxia-ischemia (HI) insult induced by left carotid artery ligation and subsequent exposure to 10% oxygen in nitrogen. The presence of ILCs and ILC2s in the brain was examined at different time points after HI. The contribution of ILC2s to HI-induced preterm brain damage was explored using a conditionally targeted ILC2-deficient mouse strain (Rorα fl/fl IL7r Cre ), and gray and white-matter injury were evaluated at 7 days post-HI. The inflammatory response in the injured brain was assessed using immunoassays and immunochemistry staining. RESULTS: Significant increases in ILCs and ILC2s were observed at 24 h, 3 days, and 7 days post-HI in the injured brain hemisphere compared with the uninjured hemisphere in wild-type mice. ILC2s in the brain were predominantly located in the meninges of the injured ipsilateral hemispheres after HI but not in the brain parenchyma. Overall, we did not observe changes in cytokine/chemokine levels in the brains of Rorα fl/fl IL7r Cre mice compared with wild type animals apart from IL-13. Gray and white-matter tissue loss in the brain was not affected after HI in Rorα fl/fl IL7r Cre mice. Correspondingly, we did not find any differences in reactive microglia and astrocyte numbers in the brain in Rorα fl/fl IL7r Cre mice compared with wild-type mice following HI insult. CONCLUSION: After HI, ILCs and ILC2s accumulate in the injured brain hemisphere. However, ILC2s do not contribute to the development of brain damage in this mouse model of preterm brain injury.

4.
Mol Psychiatry ; 24(10): 1566, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30890763

RESUMEN

Following publication of this article, the authors noticed an error in the abstract, where they incorrectly stated that: "Direct application of IL-1ß to ex vivo hippocampal slices induced non-synaptic depolarisation and irreversible loss of membrane potential in CA1 neurons from diseased animals and systemic LPS increased apoptosis in the degenerating brain, in an IL-1RI-/--dependent fashion". This has now been corrected to: "Direct application of IL-1ß to ex vivo hippocampal slices induced non-synaptic depolarisation and irreversible loss of membrane potential in CA1 neurons from diseased animals and systemic LPS increased apoptosis in the degenerating brain, in an IL-1RI-dependent fashion". The authors would like to apologise for this error. This has been corrected in both the PDF and HTML versions of the article.

5.
Glia ; 67(7): 1254-1276, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30680794

RESUMEN

Type I interferons (IFN-I) are the principal antiviral molecules of the innate immune system and can be made by most cell types, including central nervous system cells. IFN-I has been implicated in neuroinflammation during neurodegeneration, but its mechanism of induction and its consequences remain unclear. In the current study, we assessed expression of IFN-I in murine prion disease (ME7) and examined the contribution of the IFN-I receptor IFNAR1 to disease progression. The data indicate a robust IFNß response, specifically in microglia, with evidence of IFN-dependent genes in both microglia and astrocytes. This IFN-I response was absent in stimulator of interferon genes (STING-/- ) mice. Microglia showed increased numbers and activated morphology independent of genotype, but transcriptional signatures indicated an IFNAR1-dependent neuroinflammatory phenotype. Isolation of microglia and astrocytes demonstrated disease-associated microglial induction of Tnfα, Tgfb1, and of phagolysosomal system transcripts including those for cathepsins, Cd68, C1qa, C3, and Trem2, which were diminished in IFNAR1 and STING deficient mice. Microglial increases in activated cathepsin D, and CD68 were significantly reduced in IFNAR1-/- mice, particularly in white matter, and increases in COX-1 expression, and prostaglandin synthesis were significantly mitigated. Disease progressed more slowly in IFNAR1-/- mice, with diminished synaptic and neuronal loss and delayed onset of neurological signs and death but without effect on proteinase K-resistant PrP levels. Therefore, STING-dependent IFN-I influences microglial phenotype and influences neurodegenerative progression despite occurring secondary to initial degenerative changes. These data expand our mechanistic understanding of IFN-I induction and its impact on microglial function during chronic neurodegeneration.


Asunto(s)
Progresión de la Enfermedad , Interferón Tipo I/biosíntesis , Proteínas de la Membrana/deficiencia , Microglía/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Receptor de Interferón alfa y beta/deficiencia , Animales , Enfermedad Crónica , Femenino , Interferón Tipo I/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/patología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Fenotipo , Receptor de Interferón alfa y beta/genética
6.
Mol Psychiatry ; 24(10): 1533-1548, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-29875474

RESUMEN

Systemic inflammation can impair cognition with relevance to dementia, delirium and post-operative cognitive dysfunction. Episodes of delirium also contribute to rates of long-term cognitive decline, implying that these acute events induce injury. Whether systemic inflammation-induced acute dysfunction and acute brain injury occur by overlapping or discrete mechanisms remains unexplored. Here we show that systemic inflammation, induced by bacterial LPS, produces both working-memory deficits and acute brain injury in the degenerating brain and that these occur by dissociable IL-1-dependent processes. In normal C57BL/6 mice, LPS (100 µg/kg) did not affect working memory but impaired long-term memory consolidation. However prior hippocampal synaptic loss left mice selectively vulnerable to LPS-induced working memory deficits. Systemically administered IL-1 receptor antagonist (IL-1RA) was protective against, and systemic IL-1ß replicated, these working memory deficits. Dexamethasone abolished systemic cytokine synthesis and was protective against working memory deficits, without blocking brain IL-1ß synthesis. Direct application of IL-1ß to ex vivo hippocampal slices induced non-synaptic depolarisation and irreversible loss of membrane potential in CA1 neurons from diseased animals and systemic LPS increased apoptosis in the degenerating brain, in an IL-1RI-dependent fashion. The data suggest that LPS induces working memory dysfunction via circulating IL-1ß but direct hippocampal action of IL-1ß causes neuronal dysfunction and may drive neuronal death. The data suggest that acute systemic inflammation produces both reversible cognitive deficits, resembling delirium, and acute brain injury contributing to long-term cognitive impairment but that these events are mechanistically dissociable. These data have significant implications for management of cognitive dysfunction during acute illness.


Asunto(s)
Lesiones Encefálicas/inmunología , Disfunción Cognitiva/inmunología , Interleucina-1/metabolismo , Animales , Encéfalo/metabolismo , Cognición/fisiología , Trastornos del Conocimiento/inmunología , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/metabolismo , Citocinas/metabolismo , Demencia/inmunología , Femenino , Hipocampo/metabolismo , Inflamación/complicaciones , Inflamación/metabolismo , Interleucina-1/inmunología , Lipopolisacáridos/farmacología , Trastornos de la Memoria/inmunología , Memoria a Corto Plazo/fisiología , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo
7.
Front Neurol ; 9: 159, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29615958

RESUMEN

BACKGROUND: Periventricular leukomalacia (PVL) is the most common form of preterm brain injury affecting the cerebral white matter. This type of injury involves a multiphase process and is induced by many factors, including hypoxia-ischemia (HI) and infection. Previous studies have suggested that lymphocytes play a significant role in the pathogenesis of brain injury, and the aim of this study was to determine the contribution of lymphocyte subsets to preterm brain injury. METHODS: Immunohistochemistry on brain sections from neonatal mice was performed to evaluate the extent of brain injury in wild-type and T cell and B cell-deficient neonatal mice (Rag1-/- mice) using a mouse model of HI-induced preterm brain injury. Flow cytometry was performed to determine the presence of different types of immune cells in mouse brains following HI. In addition, immunostaining for CD3 T cells and CD20 B cells was performed on postmortem preterm human infant brains with PVL. RESULTS: Mature lymphocyte-deficient Rag1-/- mice showed protection from white matter loss compared to wild type mice as indicated by myelin basic protein immunostaining of mouse brains. CD3+ T cells and CD20+ B cells were observed in the postmortem preterm infant brains with PVL. Flow cytometry analysis of mouse brains after HI-induced injury showed increased frequency of CD3+ T, αßT and B cells at 7 days after HI in the ipsilateral (injured) hemisphere compared to the contralateral (control, uninjured) hemisphere. CONCLUSION: Lymphocytes were found in the injured brain after injury in both mice and humans, and lack of mature lymphocytes protected neonatal mice from HI-induced brain white matter injury. This finding provides insight into the pathology of perinatal brain injury and suggests new avenues for the development of therapeutic strategies.

8.
Am J Pathol ; 188(3): 757-767, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29248460

RESUMEN

Brain injury in premature infants, especially periventricular leukomalacia, is an important cause of neurologic disabilities. Inflammation contributes to perinatal brain injury development, but the essential mediators that lead to early-life brain injury remain largely unknown. Neonates have reduced capacity for mounting conventional αßT-cell responses. However, γδT cells are already functionally competent during early development and are important in early-life immunity. We investigated the potential contribution of γδT cells to preterm brain injury using postmortem brains from human preterm infants with periventricular leukomalacia and two animal models of preterm brain injury-the hypoxic-ischemic mouse model and a fetal sheep asphyxia model. Large numbers of γδT cells were observed in the brains of mice, sheep, and postmortem preterm infants after injury, and depletion of γδT cells provided protection in the mouse model. The common γδT-cell-associated cytokines interferon-γ and IL-17A were not detectable in the brain. Although there were increased mRNA levels of Il17f and Il22 in the mouse brains after injury, neither IL-17F nor IL-22 cytokines contributed to preterm brain injury. These findings highlight unique features of injury in the developing brain, where, unlike injury in the mature brain, γδT cells function as initiators of injury independently of common γδT-cell-associated cytokines. This finding will help to identify therapeutic targets for preventing or treating preterm infants with brain injury.


Asunto(s)
Encéfalo/patología , Hipoxia-Isquemia Encefálica/patología , Linfocitos Intraepiteliales/patología , Leucomalacia Periventricular/patología , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Hipoxia-Isquemia Encefálica/metabolismo , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Linfocitos Intraepiteliales/metabolismo , Leucomalacia Periventricular/metabolismo , Masculino , Ratones , Ovinos
9.
J Neuroinflammation ; 14(1): 255, 2017 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-29262837

RESUMEN

BACKGROUND: Infection and sepsis are associated with brain white matter injury in preterm infants and the subsequent development of cerebral palsy. METHODS: In the present study, we used a neonatal mouse sepsis-induced white matter injury model to determine the contribution of different T cell subsets (αßT cells and γδT cells) to white matter injury and consequent behavioral changes. C57BL/6J wild-type (WT), T cell receptor (TCR) δ-deficient (Tcrd -/-, lacking γδT cells), and TCRα-deficient (Tcra -/-, lacking αßT cells) mice were administered with lipopolysaccharide (LPS) at postnatal day (PND) 2. Brain myelination was examined at PNDs 12, 26, and 60. Motor function and anxiety-like behavior were evaluated at PND 26 or 30 using DigiGait analysis and an elevated plus maze. RESULTS: White matter development was normal in Tcrd -/- and Tcrα -/- compared to WT mice. LPS exposure induced reductions in white matter tissue volume in WT and Tcrα -/- mice, but not in the Tcrd -/- mice, compared with the saline-treated groups. Neither LPS administration nor the T cell deficiency affected anxiety behavior in these mice as determined with the elevated plus maze. DigiGait analysis revealed motor function deficiency after LPS-induced sepsis in both WT and Tcrα -/- mice, but no such effect was observed in Tcrd -/- mice. CONCLUSIONS: Our results suggest that γδT cells but not αßT cells contribute to sepsis-induced white matter injury and subsequent motor function abnormalities in early life. Modulating the activity of γδT cells in the early stages of preterm white matter injury might represent a novel therapeutic strategy for the treatment of perinatal brain injury.


Asunto(s)
Leucoencefalopatías/etiología , Trastornos del Movimiento/etiología , Receptores de Antígenos de Linfocitos T alfa-beta/deficiencia , Receptores de Antígenos de Linfocitos T gamma-delta/deficiencia , Sepsis/complicaciones , Animales , Animales Recién Nacidos , Ansiedad/etiología , Ansiedad/genética , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Encéfalo/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Marcha/efectos de los fármacos , Marcha/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Básica de Mielina/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Sepsis/inducido químicamente , Sepsis/patología , Bazo/patología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología
10.
Cell Biol Int ; 39(2): 224-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25077467

RESUMEN

Retinoic acid inducible gene I (RIG-I) is a well established pattern recognition receptor (PRR) in neurons infected with Japanese encephalitis virus (JEV) as reported previously from our laboratory. Japanese encephalitis (JE) virus infection in brain has been shown to decrease the proliferation of neural stem/progenitor cells (NSPCs) which has its implications in neurological sequelae in JE survivors. We have found that ablation of RIG-I both in vivo and in vitro models results in significant decrease in NSPC proliferation post JEV infection. We hypothesize that knockdown of RIG-I diminishes the expression of antiviral molecules resulting in an increase in viral replication, which in turn results in enhancement of the expression of cell cycle inhibitors, hence affecting the proliferation of NSPCs.


Asunto(s)
Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Virus de la Encefalitis Japonesa (Especie)/fisiología , Encefalitis Japonesa/patología , Encefalitis Japonesa/veterinaria , Encefalitis Japonesa/virología , Femenino , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores de Superficie Celular , Replicación Viral
11.
Biomed Res Int ; 2014: 182029, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24987671

RESUMEN

Withania somnifera (Ashwagandha), also known as Indian Ginseng, is a well-known Indian medicinal plant due to its antioxidative, antistress, antigenotoxic, and immunomodulatory properties. The present study was designed to assess and establish the cytoprotective potential of Ashwagandha leaf aqueous extract against lead induced toxicity. Pretreatment of C6 cells with 0.1% Ashwagandha extract showed cytoprotection against 25 µM to 400 µM concentration of lead nitrate. Further pretreatment with Ashwagandha extract to lead nitrate exposed cells (200 µM) resulted in normalization of glial fibrillary acidic protein (GFAP) expression as well as heat shock protein (HSP70), mortalin, and neural cell adhesion molecule (NCAM) expression. Further, the cytoprotective efficacy of Ashwagandha extract was studied in vivo. Administration of Ashwagandha extract provided significant protection to lead induced altered antioxidant defense that may significantly compromise normal cellular function. Ashwagandha also provided a significant protection to lipid peroxidation (LPx) levels, catalase, and superoxide dismutase (SOD) but not reduced glutathione (GSH) contents in brain tissue as well as peripheral organs, liver and kidney, suggesting its ability to act as a free radical scavenger protecting cells against toxic insult. These results, thus, suggest that Ashwagandha water extract may have the potential therapeutic implication against lead poisoning.


Asunto(s)
Plomo/toxicidad , Neuroglía/metabolismo , Nitratos/toxicidad , Extractos Vegetales/farmacología , Hojas de la Planta/química , Animales , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/biosíntesis , Glutatión/metabolismo , Proteínas HSP70 de Choque Térmico/biosíntesis , Masculino , Moléculas de Adhesión de Célula Nerviosa/biosíntesis , Neuroglía/patología , Oxidorreductasas/biosíntesis , Extractos Vegetales/química , Ratas , Ratas Wistar
12.
Neurobiol Dis ; 69: 235-47, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24909816

RESUMEN

Toll-like receptor 7 (TLR7) known to recognize guanidine-rich ssRNA has been shown to mount vital host defense mechanism against many viruses including flaviviruses. Signal transduction through TLR7 has been shown to produce type-1 interferon and proinflammatory mediators, thereby initiating essential innate immune response against ssRNA viruses in hosts. Systemic and brain specific TLR7 knock-down mice (TLR7(KD)) were generated using vivo-morpholinos. These mice were then subcutaneously challenged with lethal dose of JEV (GP78 strain) and were subsequently analyzed for survival. Significant difference in susceptibility to JEV between wild-type and systemic TLR7(KD) mice was observed whereas, no difference in susceptibility to JEV infection was seen in brain-specific TLR7(KD) mice. Significant decreases in IFN-α and antiviral proteins were also observed in both TLR7(KD) mice along with increased viral loads in their brain. Owing to increased viral load, increases in levels of various proinflammatory cyto/chemokines, increased microglial activation and infiltration of peripheral immune cells in brain of TLR7(KD) mice were also observed. Immunocytochemistry and RNA co-immunoprecipitation performed with JEV-infected N2a or HT22 cells indicated endosomal localization and confirmed interaction between JEV ssRNA with TLR7. Treatment of mice with imiquimod, a TLR7 agonist, prior to JEV infection resulted in their increased survival. Overall, our results suggest that the TLR7 response following JEV infection promotes type-1 interferon production and generation of antiviral state which might contribute to protective effect in systemic infection.


Asunto(s)
Encefalitis Japonesa/inmunología , Inmunidad Innata , Glicoproteínas de Membrana/metabolismo , Receptor Toll-Like 7/metabolismo , Adyuvantes Inmunológicos/farmacología , Aminoquinolinas/farmacología , Animales , Encéfalo/inmunología , Encéfalo/virología , Línea Celular , Línea Celular Tumoral , Células Cultivadas , ADN Viral/metabolismo , Modelos Animales de Enfermedad , Virus de la Encefalitis Japonesa (Especie)/genética , Encefalitis Japonesa/prevención & control , Encefalitis Japonesa/virología , Técnicas de Silenciamiento del Gen , Humanos , Imiquimod , Interferón Tipo I/metabolismo , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/genética , Ratones Endogámicos BALB C , Ratones Transgénicos , Microglía/fisiología , Neuronas/inmunología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/genética
13.
Viral Immunol ; 27(2): 48-59, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24605788

RESUMEN

An immune role of neural stem/progenitor cells (NSPCs) has been proposed in many recent studies; however much still remains to be elucidated. In the current investigation, we report that NSPCs possess the ability to convert encephalitogenic T cells into CD4(+)-CD25(+)-FOXP3(+) regulatory T cells (T(reg)). Encephalitogenic and nonencephalitogenic T cells isolated from sham and Japanese encephalitis virus (JEV) infected animals were co-cultured with mouse NSPCs. Post co-culture, significant increase in the number of T(regs) was observed from encephalitogenic T cells co-cultured with NSPCs. This increased conversion was found to be dependent on direct contact between T cells and NSPCs. Neutralization of TGF-ß and IFN-γ in NSPC cultures abrogated this increased conversion of encephalitogenic T cells into T(regs). Flow cytometric, quantitative RT-PCR, and immunoblot analysis of both T cells and NSPCs revealed surface and intracellular changes post co-culture. Co-stimulatory molecules (B7) and ICAM-1 were increased on NSPCs post co-culture; levels of TGFß, IFNγ, and TGFßR1 were also increased in NSPCs. This study provides a basic insight into the interaction between CNS-infiltrating encephalitogenic T cells and NSPCs during viral encephalitis. Conversion of encephalitogenic T cells into CD4(+)-CD25(+)-FOXP3(+) T(regs) through interaction with NSPCs indicates an attempt in regulation of excessive inflammation in the CNS.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Virus de la Encefalitis Japonesa (Especie)/inmunología , Encefalitis por Arbovirus/inmunología , Infecciones por Flavivirus/inmunología , Células-Madre Neurales/inmunología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T CD4-Positivos/química , Células Cultivadas , Técnicas de Cocultivo , Citometría de Flujo , Factores de Transcripción Forkhead/análisis , Immunoblotting , Subunidad alfa del Receptor de Interleucina-2/análisis , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena en Tiempo Real de la Polimerasa , Linfocitos T Reguladores/química
14.
Virus Res ; 185: 32-40, 2014 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-24657789

RESUMEN

The flaviviral encephalitis has now become a major health concern in global scale. The efficient detection of viral infection and induction of the innate antiviral response by host's innate immune system are crucial to determine the outcome of infection. The intracellular pattern recognition receptors TLRs, RLRs, NLRs and CLRs play a central role in detection and initiation of robust antiviral response against flaviviral infection. Both cytoplasmic RLRs, RIG-I and MDA5 have been shown to be implicated in sensing flaviviral genomic RNA. Similarly among TLRs mainly TLR3 and TLR7 are known to respond in flaviviral infections as they are known to sense dsRNA and ssRNA moiety as their natural cognate ligand. Several studies have also shown the roles of NLRs and CLRs in mounting an innate antiviral response against flavivirus but, it is yet to be completely understood. Until now only few reports have implicated NLRs and CLRs in induction of antiviral and proinflammatory state following flaviviral infection. The current review therefore aims to comprehensively analyze past as well as current understanding on the role of PRRs in flaviviral infections.


Asunto(s)
Infecciones por Flavivirus/inmunología , Flavivirus/fisiología , Receptores de Reconocimiento de Patrones/inmunología , Animales , Flavivirus/genética , Infecciones por Flavivirus/genética , Infecciones por Flavivirus/virología , Humanos , Inmunidad Innata , Receptores de Reconocimiento de Patrones/genética , Transducción de Señal
15.
Cell Immunol ; 285(1-2): 100-10, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24140964

RESUMEN

Viruses have evolved various mechanisms to subvert the host's immune system and one of them is preventing the infected cells from sending out chemotactic signals to activate the adaptive immune response. Japanese encephalitis virus (JEV) is a neuropathologic flavivirus that is responsible for significant number of child mortalities in various parts of South-East Asia. In this study we show that JEV modulates suppressors of cytokine signaling (SOCS)1 and 3 expression in macrophages to bring about changes in the JAK-STAT signaling cascade, so as to inhibit proinflammatory cyto/chemokine release. Using real time PCR, immunoblotting and immunofluorescent staining, we show that the expression of type 1 interferons and intracellular expression of viral genes are also affected over time. Also, following the initial activation of SOCS1 and 3, there is production of interferon-inducible anti-viral proteins in the cells which may be responsible for inhibiting viral replication. However, even at later time points, viral genes were still detected from the macrophages, albeit at lesser quantities, than earlier time points, indicative of intracellular persistence of the virus in a latent form. On knocking down SOCS1 and SOCS3 we found a significant decrease in viral gene expression at an early time point, indicating the dysregulation of the signaling cascade leading to increased production of interferon-inducible anti-viral proteins. Taken together, our study provides an insight into the role of JEV infection in modulating the JAK-STAT pathway with the help of SOCS leading to the generation of an antiviral innate immune response.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/metabolismo , Encefalitis Japonesa/inmunología , Macrófagos/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Células Cultivadas , Quimiocinas/biosíntesis , Citocinas/biosíntesis , Virus de la Encefalitis Japonesa (Especie)/inmunología , Femenino , Inmunidad Innata/inmunología , Interferón Tipo I/biosíntesis , Quinasas Janus/metabolismo , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , FN-kappa B/antagonistas & inhibidores , Interferencia de ARN , ARN Interferente Pequeño , Factores de Transcripción STAT/metabolismo , Transducción de Señal/inmunología , Proteína 1 Supresora de la Señalización de Citocinas , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/biosíntesis , Proteínas Supresoras de la Señalización de Citocinas/genética , Replicación Viral/inmunología
16.
Sci Rep ; 2: 347, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22470840

RESUMEN

Flavivirus-mediated inflammation causes neuronal death, but whether the infected neurons can evoke an innate immune response to elicit their own protection, is unknown. In an earlier study we have shown that neuronal RIG-I, play a significant role in inducing production and release of molecules that are related to inflammation. In this study, using a neuronal cell line, we show that RIG-I acts with STING in a concerted manner following its interaction with Japanese encephalitis viral RNA to induce a type 1 interferon response. Knock-down of STING showed that the expressions of various inflammatory signaling molecules were down-regulated along with increased intracellular viral load. Alternatively, over-expressing STING decreased intracellular viral load. Our results indicate that at the sub-cellular level, interaction between the pattern recognition receptor RIG-I and the adapter molecule STING, is a major contributor to elicit immunological responses involving the type 1 interferons in neurons following JEV infections.


Asunto(s)
Encefalitis Japonesa/inmunología , Inmunidad Innata/fisiología , Proteínas de la Membrana/fisiología , Neuronas/inmunología , Ciclooxigenasa 2/metabolismo , Virus de la Encefalitis Japonesa (Especie)/genética , Humanos , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/metabolismo , FN-kappa B/metabolismo , Regulación hacia Arriba , Carga Viral , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
PLoS One ; 6(6): e21761, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21738791

RESUMEN

BACKGROUND: Neuroinflammation associated with Japanese encephalitis (JE) is mainly due to the activation of glial cells with subsequent release of proinflammatory mediators from them. The recognition of viral RNA, in part, by the pattern recognition receptor retinoic acid-inducible gene I (RIG-I) has been indicated to have a role in such processes. Even though neurons are also known to express this receptor, its role after JE virus (JEV) infections is yet to be elucidated. METHODOLOGY/PRINCIPAL FINDINGS: Upon infecting murine neuroblastoma cells and primary cortical neurons with JEV the expression profile of key proinflammatory cyto/chemokines were analyzed by qRT-PCR and bead array, both before and after ablation of RIG-I. Immunoblotting was performed to evaluate the levels of key molecules downstream to RIG-I leading to production of proinflammatory mediators. Changes in the intracellular viral antigen expression were confirmed by intracellular staining and immunoblotting. JEV infection induced neuronal expression of IL-6, IL-12p70, MCP-1, IP-10 and TNF-α in a time-dependent manner, which showed significant reduction upon RIG-I ablation. Molecules downstream to RIG-I showed significant changes upon JEV-infection, that were modulated following RIG-I ablation. Ablation of RIG-I in neurons also increased their susceptibility to JEV. CONCLUSIONS/SIGNIFICANCE: In this study we propose that neurons are one of the potential sources of proinflammatory cyto/chemokines in JEV-infected brain that are produced via RIG-I dependent pathways. Ablation of RIG-I in neurons leads to increased viral load and reduced release of the cyto/chemokines.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Virus de la Encefalitis Japonesa (Especie)/fisiología , Inmunidad Innata/fisiología , Neuronas/metabolismo , Neuronas/virología , Animales , Línea Celular , Línea Celular Tumoral , Quimiocina CCL2/metabolismo , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , Inmunidad Innata/genética , Interleucina-12/metabolismo , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos BALB C , Factor de Necrosis Tumoral alfa/metabolismo
18.
Infect Disord Drug Targets ; 11(3): 300-14, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21488831

RESUMEN

Chemotherapy in Japanese encephalitis (JE) is at present entirely supportive and not targeted at the virus. There are no available drugs to effectively counter the viral infection, thereby making the fight against JE a daunting task. With approximately 50,000 reported cases per year, nearly 10,000 deaths and 3 billion people living in endemic regions, it is imperative that the hunt for an effective drug be expedited. Prophylactic measures are effective against JE, but the problem plaguing all is the underdevelopment and inefficiency of medical services in developing countries. Combined to that are difficulties to earn a living and illiteracy, that leaves significant proportions of the population in these countries uninformed about the magnitude of the threat and uninterested in the potential benefits of prophylactic strategies. Thus, for such countries coming under the JE endemic region, the need for developing therapeutic strategies that are cheap, easily available and with no or tolerable side effects, becomes significant. With rapid globalization and a gradual shift in global climate, JEV, like many other flaviviruses, may emerge in newer areas. This review is an effort to briefly outline the chemotherapeutic approaches adopted over the years in developing effective therapeutic countermeasures against this deadly disease and highlights the promising avenues that need to be treaded in order to win the war against JEV.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie) , Encefalitis Japonesa/tratamiento farmacológico , Animales , Países en Desarrollo , Encefalitis Japonesa/prevención & control , Encefalitis Japonesa/virología , Humanos , Insectos Vectores/virología , Vacunas contra la Encefalitis Japonesa/administración & dosificación , Zoonosis/transmisión , Zoonosis/virología
19.
J Neuroimmune Pharmacol ; 6(3): 420-33, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21424747

RESUMEN

Inflammation in the central nervous system (CNS) in Japanese encephalitis (JE) is shown to be the result of microglial activation that leads to the release of various proinflammatory mediators. Peripheral macrophages have been reported to infiltrate into the CNS in JE, though their contribution to the inflammatory process is yet to be elucidated. In this study, using an in vitro macrophage model, we have shown that upon JE virus infection, these cells secrete various soluble factors which may significantly add to the existing inflammatory milieu and lead to apoptotic or necrotic death of neurons. However, it is difficult to quantify the extent of involvement of either the microglia or infiltrating macrophages in the inflammatory processes.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie) , Encefalitis Japonesa/inmunología , Macrófagos/inmunología , Macrófagos/virología , Neuronas/patología , Animales , Muerte Celular , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Immunoblotting , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Neuronas/inmunología , Especies Reactivas de Oxígeno/metabolismo
20.
PLoS Negl Trop Dis ; 4(11): e892, 2010 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-21124882

RESUMEN

BACKGROUND: Japanese encephalitis (JE), caused by a mosquito-borne flavivirus, is endemic to the entire south-east Asian and adjoining regions. Currently no therapeutic interventions are available for JE, thereby making it one of the most dreaded encephalitides in the world. An effective way to counter the virus would be to inhibit viral replication by using anti-sense molecules directed against the viral genome. Octaguanidinium dendrimer-conjugated Morpholino (or Vivo-Morpholino) are uncharged anti-sense oligomers that can enter cells of living organisms by endocytosis and subsequently escape from endosomes into the cytosol/nuclear compartment of cells. We hypothesize that Vivo-Morpholinos generated against specific regions of 3' or 5' untranslated regions of JEV genome, when administered in an experimental model of JE, will have significant antiviral and neuroprotective effect. METHODOLOGY/PRINCIPAL FINDINGS: Mice were infected with JEV (GP78 strain) followed by intraperitoneal administration of Morpholinos (5 mg/kg body weight) daily for up to five treatments. Survivability of the animals was monitored for 15 days (or until death) following which they were sacrificed and their brains were processed either for immunohistochemical staining or protein extraction. Plaque assay and immunoblot analysis performed from brain homogenates showed reduced viral load and viral protein expression, resulting in greater survival of infected animals. Neuroprotective effect was observed by thionin staining of brain sections. Cytokine bead array showed reduction in the levels of proinflammatory cytokines in brain following Morpholino treatment, which were elevated after infection. This corresponded to reduced microglial activation in brain. Oxidative stress was reduced and certain stress-related signaling molecules were found to be positively modulated following Morpholino treatment. In vitro studies also showed that there was decrease in infective viral particle production following Morpholino treatment. CONCLUSIONS/SIGNIFICANCE: Administration of Vivo-Morpholino effectively resulted in increased survival of animals and neuroprotection in a murine model of JE. Hence, these oligomers represent a potential antiviral agent that merits further evaluation.


Asunto(s)
Antivirales/administración & dosificación , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Encefalitis Japonesa/tratamiento farmacológico , Morfolinas/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Oligonucleótidos/administración & dosificación , Animales , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/virología , Chlorocebus aethiops , Citocinas/inmunología , Dendrímeros/administración & dosificación , Virus de la Encefalitis Japonesa (Especie)/genética , Virus de la Encefalitis Japonesa (Especie)/fisiología , Encefalitis Japonesa/inmunología , Encefalitis Japonesa/prevención & control , Encefalitis Japonesa/virología , Humanos , Ratones , Ratones Endogámicos BALB C , Oligodesoxirribonucleótidos Antisentido/administración & dosificación , Oligodesoxirribonucleótidos Antisentido/genética , Oligonucleótidos/genética , Células Vero , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...