Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros











Intervalo de año de publicación
1.
Int J Mol Sci ; 23(19)2022 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-36232593

RESUMEN

Type I interferons (IFN), including IFNß, play a protective role in multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). Type I IFNs are induced by the stimulation of innate signaling, including via cytoplasmic RIG-I-like receptors. In the present study, we investigated the potential effect of a chimeric protein containing the key domain of RIG-I signaling in the production of CNS endogenous IFNß and asked whether this would exert a therapeutic effect against EAE. We intrathecally administered an adeno-associated virus vector (AAV) encoding a fusion protein comprising RIG-I 2CARD domains (C) and the first 200 amino acids of mitochondrial antiviral-signaling protein (MAVS) (M) (AAV-CM). In vivo imaging in IFNß/luciferase reporter mice revealed that a single intrathecal injection of AAV-CM resulted in dose-dependent and sustained IFNß expression within the CNS. IFNß expression was significantly increased for 7 days. Immunofluorescent staining in IFNß-YFP reporter mice revealed extraparenchymal CD45+ cells, choroid plexus, and astrocytes as sources of IFNß. Moreover, intrathecal administration of AAV-CM at the onset of EAE induced the suppression of EAE, which was IFN-I-dependent. These findings suggest that accessing the signaling pathway downstream of RIG-I represents a promising therapeutic strategy for inflammatory CNS diseases, such as MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Interferón Tipo I , Aminoácidos , Animales , Antivirales , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Interferón Tipo I/metabolismo , Interferón beta/genética , Interferón beta/metabolismo , Ratones , Proteínas Recombinantes de Fusión , Transducción de Señal
2.
Int J Mol Sci ; 23(16)2022 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-36012246

RESUMEN

Recently, the protective and/or pathological role of virus-specific T cells in SARS-CoV-2 infection has been the focus of many studies. We investigated the anti-spike IgG levels and SARS-CoV-2-specific T cells in 125 donors (90 vaccinated with four different vaccine platforms, 16 individuals with a previous natural infection, and 19 not vaccinated donors who did not report previous SARS-CoV-2 infections). Our data show that anti-spike IgG titers were similar between naturally infected subjects and those vaccinated with adenoviral vector vaccines. Of note, all immunized donors produced memory CD4+ and/or CD8+ T cells. A sustained polyfunctionality of SARS-CoV-2-specific T cells in all immunized donors was also demonstrated. Altogether, our data suggest that the natural infection produces an overall response like that induced by vaccination. Therefore, this detailed immunological evaluation may be relevant for other vaccine efforts especially for the monitoring of novel vaccines effective against emerging virus variants.


Asunto(s)
COVID-19 , Vacunas Virales , Anticuerpos Antivirales , Linfocitos T CD8-positivos , COVID-19/prevención & control , Humanos , Inmunoglobulina G , SARS-CoV-2 , Vacunación
3.
Stem Cell Reports ; 16(2): 281-294, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33482100

RESUMEN

Microglia have recently been established as key regulators of brain development. However, their role in neuronal subtype specification remains largely unknown. Using three different co-culture setups, we show that microglia-secreted factors enhance dopaminergic differentiation of somatic and induced pluripotent stem cell-derived human neural stem cells (NSCs). The effect was consistent across different NSC and microglial cell lines and was independent of prior microglial activation, although restricted to microglia of embryonic origin. We provide evidence that the effect is mediated through reduced cell proliferation and decreased apoptosis and necrosis orchestrated in a sequential manner during the differentiation process. tumor necrosis factor alpha, interleukin-1ß, and insulinlike growth factor 1 are identified as key mediators of the effect and shown to directly increase dopaminergic differentiation of human NSCs. These findings demonstrate a positive effect of microglia on dopaminergic neurogenesis and may provide new insights into inductive and protective factors that can stimulate in vitro derivation of dopaminergic neurons.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Citocinas/metabolismo , Neuronas Dopaminérgicas/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Microglía/fisiología , Células-Madre Neurales/metabolismo , Animales , Apoptosis , Línea Celular , Células Cultivadas , Técnicas de Cocultivo/métodos , Dopamina/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Neurogénesis , Factor de Necrosis Tumoral alfa/metabolismo
4.
Scand J Immunol ; 92(5): e12963, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32851668

RESUMEN

Myeloid cells represent the major cellular component of innate immune responses. Myeloid cells include monocytes and macrophages, granulocytes (neutrophils, basophils and eosinophils) and dendritic cells (DC). The role of myeloid cells has been broadly described both in physiological and in pathological conditions. All tissues or organs are equipped with resident myeloid cells, such as parenchymal microglia in the brain, which contribute to maintaining homeostasis. Moreover, in case of infection or tissue damage, other myeloid cells such as monocytes or granulocytes (especially neutrophils) can be recruited from the circulation, at first to promote inflammation and later to participate in repair and regeneration. This review aims to address the regulatory roles of myeloid cells in inflammatory diseases of the central nervous system (CNS), with a particular focus on recent work showing induction of suppressive function via stimulation of innate signalling in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE).


Asunto(s)
Sistema Nervioso Central/inmunología , Células Dendríticas/inmunología , Granulocitos/inmunología , Inflamación/inmunología , Macrófagos/inmunología , Células Mieloides/inmunología , Animales , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Humanos , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología
5.
Front Immunol ; 11: 1110, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32582192

RESUMEN

Background: MiR-146a is an important regulator of innate inflammatory responses and is also implicated in cell death and survival. Methods: By sorting CNS resident cells, microglia were the main cellular source of miR-146a. Therefore, we investigated microglia function and phenotype in miR-146a knock-out (KO) mice, analyzed the proteome of KO and wild-type (WT) microglia by LC-MS/MS, and examined miR-146a expression in different brain lesions of patients with multiple sclerosis (MS). Results: When stimulated with LPS or myelin in vitro, microglia from KO mice expressed higher levels of IL-1ß, TNF, IL-6, IL-10, CCL3, and CCL2 compared to WT. Stimulation increased migration and phagocytosis of WT but not KO microglia. CD11c+ microglia were induced by cuprizone (CPZ) in the WT mice but less in the KO. The proteome of ex vivo microglia was not different in miR-146a KO compared to WT mice, but CPZ treatment induced differential and reduced protein responses in the KO: GOT1, COX5b, CRYL1, and cystatin-C were specifically changed in KO microglia. We explored discriminative features of microglia proteomes: sparse Partial Least Squares-Discriminant Analysis showed the best discrimination when control and CPZ-treated conditions were compared. Cluster of ten proteins separated WT and miR-146a KO microglia after CPZ: among them were sensomes allowing to perceive the environment, Atp1a3 that belongs to the signature of CD11c+ microglia, and proteins related to inflammatory responses (S100A9, Ppm1g). Finally, we examined the expression of miR-146a and its validated target genes in different brain lesions of MS patients. MiR-146 was upregulated in all lesion types, and the highest expression was in active lesions. Nineteen of 88 validated target genes were significantly changed in active lesions, while none were changed in NAWM. Conclusion: Our data indicated that microglia is the major source of miR-146a in the CNS. The absence of miR-146a differentially affected microglia function and proteome, and miR-146a may play an important role in gene regulation of active MS lesions.


Asunto(s)
MicroARNs/metabolismo , Microglía/metabolismo , Esclerosis Múltiple/metabolismo , Animales , Quelantes/toxicidad , Cuprizona/toxicidad , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/inmunología , Microglía/inmunología , Esclerosis Múltiple/inmunología , Proteoma
6.
Mult Scler ; 26(10): 1187-1196, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-31287367

RESUMEN

BACKGROUND: Neuromyelitis optica spectrum disorder (NMOSD) is a relapsing inflammatory central nervous system (CNS) disease for which there is no cure. Immunoglobulin G autoantibodies specific for the water channel aquaporin-4 are a serum biomarker, believed to induce complement-dependent astrocyte damage with secondary demyelination. OBJECTIVE: To investigate the effect of angiotensin AT2 receptor (AT2R) stimulation on NMOSD-like pathology and its underlying mechanism. METHODS: NMOSD-like pathology was induced in mice by intracerebral injection of immunoglobulin-G isolated from NMOSD patient serum, with complement. This mouse model produces the characteristic histological features of NMOSD. A specific AT2R agonist, Compound 21 (C21), was given intracerebrally at day 0 and by intrathecal injection at day 2. RESULTS: Loss of aquaporin-4 and glial fibrillary acidic protein was attenuated by treatment with C21. Administration of C21 induced mRNA for interleukin-10 in the brain. NMOSD-like pathology was exacerbated in interleukin-10-deficient mice, suggesting a protective role. C21 treatment did not attenuate NMOSD-like pathology in interleukin-10-deficient mice, indicating that the protective effect of AT2R stimulation was dependent on interleukin-10. CONCLUSION: Our findings identify AT2R as a novel potential therapeutic target for the treatment of NMOSD. Interleukin-10 signaling is an essential part of the protective mechanism counteracting NMOSD pathology.


Asunto(s)
Neuromielitis Óptica , Animales , Acuaporina 4/genética , Autoanticuerpos , Humanos , Interleucina-10 , Ratones , Recurrencia Local de Neoplasia , Neuromielitis Óptica/tratamiento farmacológico , Receptor de Angiotensina Tipo 2
7.
Front Immunol ; 9: 490, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29593734

RESUMEN

Background: The cuprizone (CPZ) model of multiple sclerosis (MS) was used to identify microRNAs (miRNAs) related to in vivo de- and remyelination. We further investigated the role of miR-146a in miR-146a-deficient (KO) mice: this miRNA is differentially expressed in MS lesions and promotes differentiation of oligodendrocyte precursor cells (OPCs) during remyelination, but its role has not been examined during demyelination. Methods: MicroRNAs were examined by Agilent Mouse miRNA Microarray in the corpus callosum during CPZ-induced demyelination and remyelination. Demyelination, axonal loss, changes in number of oligodendrocytes, OPCs, and macrophages/microglia was compared by histology/immunohistochemistry between KO and WT mice. Differential expression of target genes and proteins of miR-146a was analyzed in the transcriptome (4 × 44K Agilent Whole Mouse Genome Microarray) and proteome (liquid chromatography tandem mass spectrometry) of CPZ-induced de- and remyelination in WT mice. Levels of proinflammatory molecules in the corpus callosum were compared in WT versus KO mice by Meso Scale Discovery multiplex protein analysis. Results: miR-146a was increasingly upregulated during CPZ-induced de- and remyelination. The absence of miR-146a in KO mice protected against demyelination, axonal loss, body weight loss, and atrophy of thymus and spleen. The number of CNP+ oligodendrocytes was increased during demyelination in the miR-146a KO mice, while there was a trend of increased number of NG2+ OPCs in the WT mice. miR-146a target genes, SNAP25 and SMAD4, were downregulated in the proteome of demyelinating corpus callosum in WT mice. Higher levels of SNAP25 were measured by ELISA in the corpus callosum of miR-146a KO mice, but there was no difference between KO and WT mice during demyelination. Multiplex protein analysis of the corpus callosum lysate revealed upregulated TNF-RI, TNF-RII, and CCL2 in the WT mice in contrast to KO mice. The number of Mac3+ and Iba1+ macrophages/microglia was reduced in the demyelinating corpus callosum of the KO mice. Conclusion: During demyelination, absence of miR-146a reduced inflammatory responses, demyelination, axonal loss, the number of infiltrating macrophages, and increased the number of myelinating oligodendrocytes. The number of OPCs was slightly higher in the WT mice during remyelination, indicating a complex role of miR-146a during in vivo de- and remyelination.


Asunto(s)
Axones/patología , Cuerpo Calloso/fisiología , Enfermedades Desmielinizantes/genética , MicroARNs/genética , Oligodendroglía/fisiología , Animales , Diferenciación Celular , Quimiocina CCL2/genética , Cuprizona , Femenino , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Receptores del Factor de Necrosis Tumoral/genética
8.
APMIS ; 125(11): 945-956, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28836736

RESUMEN

CCL2 is a chemokine that can be induced during neuroinflammation to recruit immune cells, but its role in the central nervous system (CNS) is unclear. Our aim was to better understand its role. We induced CCL2 in CNS of naive CCL2-deficient mice using intrathecally administered replication-defective adenovirus and examined cell infiltration by flow cytometry. CCL2 expression induced pronounced and unexpected recruitment of regulatory and IFNγ-producing T cells to CNS from blood, possibly related to defective egress of monocytes from CCL2-deficient bone marrow. Infiltration also occurred in mice lacking CCR2, a receptor for CCL2. Expression of another receptor for CCL2, CCR4, and CXCR3, a receptor for CXCL10, which was also induced, were both increased in CCL2-treated CNS. CCR4 was expressed by neurons and astrocytes as well as CD4 T cells, and CXCR3 was expressed by CD4 and CD8 T cells. Chemokine-recruited T cells did not lead to CNS pathology. Our findings show a role for CCL2 in recruitment of CD4 T cells to the CNS and show that redundancy among chemokine receptors ensures optimal response.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Sistema Nervioso Central/inmunología , Quimiocina CCL2/inmunología , Receptores CCR2/inmunología , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Astrocitos/citología , Astrocitos/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Movimiento Celular , Sistema Nervioso Central/citología , Quimiocina CCL2/deficiencia , Quimiocina CCL2/genética , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Femenino , Regulación de la Expresión Génica , Genes Reporteros , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Inyecciones Espinales , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Noqueados , Neuronas/citología , Neuronas/inmunología , Receptores CCR2/genética , Receptores CCR4/genética , Receptores CCR4/inmunología , Receptores CXCR3/genética , Receptores CXCR3/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Transducción de Señal , Proteína Fluorescente Roja
9.
J Neuroinflammation ; 11: 57, 2014 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-24666681

RESUMEN

BACKGROUND: Tissue-resident antigen-presenting cells (APC) exert a major influence on the local immune environment. Microglia are resident myeloid cells in the central nervous system (CNS), deriving from early post-embryonic precursors, distinct from adult hematopoietic lineages. Dendritic cells (DC) and macrophages infiltrate the CNS during experimental autoimmune encephalomyelitis (EAE). Microglia are not considered to be as effective APC as DC or macrophages. METHODS: In this work we compared the antigen presenting capacity of CD11c⁺ and CD11c⁻ microglia subsets with infiltrating CD11c⁺ APC, which include DC. The microglial subpopulations (CD11c⁻ CD45dim CD11b⁺ and CD11c⁺ CD45dim CD11b⁺) as well as infiltrating CD11c⁺ CD45high cells were sorted from CNS of C57BL/6 mice with EAE. Sorted cells were characterised by flow cytometry for surface phenotype and by quantitative real-time PCR for cytokine expression. They were co-cultured with primed T cells to measure induction of T cell proliferation and cytokine response. RESULTS: The number of CD11c⁺ microglia cells increased dramatically in EAE. They expressed equivalent levels of major histocompatibility complex and co-stimulatory ligands CD80 and CD86 as those expressed by CD11c⁺ cells infiltrating from blood. CD11c⁺ microglia differed significantly from blood-derived CD11c⁺ cells in their cytokine profile, expressing no detectable IL-6, IL-12 or IL-23, and low levels of IL-1ß. By contrast, CD11c⁻ microglia expressed low but detectable levels of all these cytokines. Transforming growth factor ß expression was similar in all three populations. Although CNS-resident and blood-derived CD11c⁺ cells showed equivalent ability to induce proliferation of myelin oligodendrocyte glycoprotein-immunised CD4⁺ T cells, CD11c⁺ microglia induced lower levels of T helper (Th)1 and Th17 cytokines, and did not induce Th2 cytokines. CONCLUSIONS: Our findings show distinct subtypes of APC in the inflamed CNS, with a hierarchy of functional competence for induction of CD4⁺ T cell responses.


Asunto(s)
Células Presentadoras de Antígenos/metabolismo , Proliferación Celular , Sistema Nervioso Central/patología , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Microglía/metabolismo , Linfocitos T/fisiología , Animales , Antígenos CD/metabolismo , Antígeno CD11c/metabolismo , Sistema Nervioso Central/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Citometría de Flujo , Complejo Mayor de Histocompatibilidad/fisiología , Ratones , Ratones Endogámicos C57BL
10.
PLoS One ; 7(4): e35066, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22509382

RESUMEN

Pulmonary surfactant protein D (SP-D) is a host defence lectin of the innate immune system that enhances clearance of pathogens and modulates inflammatory responses. Recently it has been found that systemic SP-D is associated with metabolic disturbances and that SP-D deficient mice are mildly obese. However, the mechanism behind SP-D's role in energy metabolism is not known.Here we report that SP-D deficient mice had significantly higher ad libitum energy intake compared to wild-type mice and unchanged energy expenditure. This resulted in accumulation but also redistribution of fat tissue. Blood pressure was unchanged. The change in energy intake was unrelated to the basal levels of hypothalamic Pro-opiomelanocortin (POMC) and Agouti-related peptide (AgRP) gene expression. Neither short time systemic, nor intracereberoventricular SP-D treatment altered the hypothalamic signalling or body weight accumulation.In ad libitum fed animals, serum leptin, insulin, and glucose were significantly increased in mice deficient in SP-D, and indicative of insulin resistance. However, restricted diets eliminated all metabolic differences except the distribution of body fat. SP-D deficiency was further associated with elevated levels of systemic bacterial lipopolysaccharide.In conclusion, our findings suggest that lack of SP-D mediates modulation of food intake not directly involving hypothalamic regulatory pathways. The resulting accumulation of adipose tissue was associated with insulin resistance. The data suggest SP-D as a regulator of energy intake and body composition and an inhibitor of metabolic endotoxemia. SP-D may play a causal role at the crossroads of inflammation, obesity, and insulin resistance.


Asunto(s)
Tejido Adiposo/metabolismo , Metabolismo Energético/genética , Pulmón/metabolismo , Proteína D Asociada a Surfactante Pulmonar/genética , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Animales , Glucemia , Sistema Nervioso Central/efectos de los fármacos , Endotoxemia/genética , Ácidos Grasos no Esterificados/sangre , Hiperfagia/genética , Hiperfagia/metabolismo , Inmunidad Innata/efectos de los fármacos , Insulina/sangre , Resistencia a la Insulina/genética , Leptina/sangre , Ratones , Proteína D Asociada a Surfactante Pulmonar/administración & dosificación , Proteína D Asociada a Surfactante Pulmonar/deficiencia
11.
Glia ; 60(4): 630-8, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22271465

RESUMEN

Within the central nervous system, astrocytes and microglia are the primary responders to endogenous ligands released upon injury and stress, as well as to infectious pathogens. Toll-like receptors (TLRs) are implicated in recognition of both types of stimulus. Whether astrocytes respond as strongly as microglia to TLR agonists remains contentious. In this study, we have rigorously purified astrocytes to determine their capacity for autonomous TLR response, in absence of microglia. We used flow cytometry and differential adhesion as well as a myeloid lineage-specific suicide gene to purify astrocytes from mixed glial cultures and measured their response to TLR agonists. Our results show that the response of astrocytes to TLR2 and TLR3 agonists is greatly enhanced by, and response to TLR4 agonists is completely dependent on, the presence of functional microglia. In the case of the TLR4 response to lipopolysaccharide, microglia exert their effect on astrocytes at least partially through release of soluble mediators that directly activate or facilitate astrocyte responses. Our findings underline the contribution of glial crosstalk in CNS responses to injury or inflammation.


Asunto(s)
Astrocitos/metabolismo , Microglía/fisiología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Animales Recién Nacidos , Antígenos CD/metabolismo , Astrocitos/efectos de los fármacos , Encéfalo/citología , Antígeno CD11b/genética , Células Cultivadas , Quimiocina CCL2/metabolismo , Técnicas de Cocultivo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Proteína Ácida Fibrilar de la Glía/metabolismo , Glutamina/análogos & derivados , Glutamina/farmacología , Inductores de Interferón/farmacología , Interleucina-1beta/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Compuestos Organometálicos/farmacología , Poli I-C/farmacología , Polisacáridos/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
12.
J Neuroimmunol ; 224(1-2): 45-50, 2010 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-20537410

RESUMEN

Chemokines are critical mediators of immune cell entry into the central nervous system (CNS), as occurs in neuroinflammatory disease such as multiple sclerosis. Chemokines are also implicated in the immune response to viral infections. Many viruses encode proteins that mimic or block chemokine actions, in order to evade host immune responses. The murine gammaherpesvirus-68 encodes a chemokine-binding protein called M3, which has unique biochemical features that enable it to bind to and inhibit an unusually broad range of chemokines. We applied a replication-defective adenoviral vector encoding M3 (AdM3) directly to the CNS to evaluate the capacity of this protein to inhibit neuroinflammation using the experimental autoimmune encephalomyelitis (EAE) model. Treatment with the AdM3 vector significantly reduced the clinical severity of EAE, attenuated CNS histopathology, and reduced numbers of immune cells infiltrating the CNS. These results suggest that M3 may represent a novel therapeutic approach to neuroinflammatory disease.


Asunto(s)
Quimiocinas/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Rhadinovirus/inmunología , Proteínas Virales/uso terapéutico , Adenoviridae/genética , Animales , Línea Celular , Inhibición de Migración Celular/genética , Inhibición de Migración Celular/inmunología , Quimiocinas/antagonistas & inhibidores , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/virología , Femenino , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Humanos , Ratones , Ratones Endogámicos C57BL , Unión Proteica/inmunología , Rhadinovirus/genética , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Proteínas Virales/genética
13.
Hematol Oncol ; 28(3): 142-50, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20155735

RESUMEN

Diffuse large B-cell lymphoma (DLBCL) is an aggressive malignancy with a variable response to therapy. We have previously shown that DLBCL cell lines differ in their susceptibility to CD40-mediated cell death, and that resistance to CD40-targeted antibodies correlated with increased expression of markers of immature B-cell and absence of Vav-1 mRNA. We used gene expression profiling to investigate the mechanism of CD40 resistance in these cell lines, and found that resistance correlated with lack of Vav-1 and inability to activate NFκB upon CD40 ligation. Analysis of tissue microarrays of 213 DLBCL cases revealed that Vav-1 expression correlated with a higher proliferative index and the presence of the post-germinal centre marker Irf-4. Our results suggest that Vav-1 expression may be associated with activated B-cell DLBCL origin and higher proliferative activity, and indicate Vav-1 as a potential marker to identify tumours likely to respond to CD40-targeted therapies.


Asunto(s)
Antígenos CD40/metabolismo , Linfoma de Células B Grandes Difuso/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-vav/biosíntesis , Anciano , Antígenos CD40/genética , Antígenos CD40/inmunología , Muerte Celular/fisiología , Línea Celular Tumoral , Activación Enzimática , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Factores Reguladores del Interferón/biosíntesis , Factores Reguladores del Interferón/metabolismo , Antígeno Ki-67/biosíntesis , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/inmunología , Linfoma de Células B Grandes Difuso/patología , Masculino , Persona de Mediana Edad , FN-kappa B/inmunología , Pronóstico , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Proto-Oncogénicas c-vav/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética
14.
Am J Pathol ; 174(5): 1880-90, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19349355

RESUMEN

Remyelination of lesions in the central nervous system contributes to neural repair following clinical relapses in multiple sclerosis. Remyelination is initiated by recruitment and differentiation of oligodendrocyte progenitor cells (OPCs) into myelinating oligodendrocytes. Simvastatin, a blood-brain barrier-permeable statin in multiple sclerosis clinical trials, has been shown to impact the in vitro processes that have been implicated in remyelination. Animals were fed a cuprizone-supplemented diet for 6 weeks to induce localized demyelination in the corpus callosum; subsequent return to normal diet for 3 weeks stimulated remyelination. Simvastatin was injected intraperitoneally during the period of coincident demyelination and OPC maturation (weeks 4 to 6), throughout the entire period of OPC responses (weeks 4 to 9), or during the remyelination-only phase (weeks 7 to 9). Simvastatin treatment (weeks 4 to 6) caused a decrease in myelin load and both Olig2(strong) and Nkx2.2(strong) OPC numbers. Simvastatin treatment (weeks 4 to 9 and 7 to 9) caused a decrease in myelin load, which was correlated with a reduction in Nkx2.2(strong) OPCs and an increase in Olig2(strong) cells, suggesting that OPCs were maintained in an immature state (Olig2(strong)/Nkx2.2(weak)). NogoA+ oligodendrocyte numbers were decreased during all simvastatin treatment regimens. Our findings suggest that simvastatin inhibits central nervous system remyelination by blocking progenitor differentiation, indicating the need to monitor effects of systemic immunotherapies that can access the central nervous system on brain tissue-repair processes.


Asunto(s)
Anticolesterolemiantes/farmacología , Enfermedades Desmielinizantes/tratamiento farmacológico , Vaina de Mielina/metabolismo , Regeneración Nerviosa/fisiología , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Simvastatina/farmacología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Western Blotting , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quelantes/administración & dosificación , Cuprizona/administración & dosificación , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/fisiología , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vaina de Mielina/patología , Proteínas del Tejido Nervioso/fisiología , Factor de Transcripción 2 de los Oligodendrocitos , Oligodendroglía/metabolismo , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Factores de Transcripción/fisiología , Proteínas de Pez Cebra
15.
J Immunol ; 181(9): 6481-90, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18941239

RESUMEN

Injury to the CNS provokes an innate inflammatory reaction that engages infiltrating leukocytes with the capacity to repair and/or exacerbate tissue damage. The initial cues that orchestrate leukocyte entry remain poorly defined. We have used flow cytometry to investigate whether MyD88, an adaptor protein that transmits signals from TLRs and receptors for IL-1 and IL-18, regulates leukocyte infiltration into the stab-injured entorhinal cortex (EC) and into sites of axonal degeneration in the denervated hippocampus. We have previously established the kinetics of leukocyte entry into the denervated hippocampus. We now show that significant leukocyte entry into the EC occurs within 3-12 h of stab injury. Whereas T cells showed small, gradual increases over 8 days, macrophage infiltration was pronounced and peaked within 12-24 h. MyD88 deficiency significantly reduced macrophage and T cell recruitment to the stab-injured EC and the denervated hippocampus at 5 days post-injury. Whereas macrophage and T cell entry remained impaired into the denervated hippocampus of MyD88-deficient mice at 8 days, leukocyte infiltration into the stab-injured EC was restored to levels observed in wild-type mice. Transcripts for TNF-alpha, IL-1beta, and CCL2, which increased >50-fold after stab injury in C57BL/6 mice at the time of peak expression, were severely reduced in injured MyD88 knockout mice. Leukocyte recruitment and gene expression were unaffected in TLR2-deficient or TLR4 mutant mice. No significant differences in gene expression were observed in mice lacking IL-1R or IL-18R. These data show that MyD88-dependent signaling mediates proinflammatory gene expression and leukocyte recruitment after CNS injury.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Corteza Entorrinal/inmunología , Corteza Entorrinal/patología , Mediadores de Inflamación/fisiología , Factor 88 de Diferenciación Mieloide/fisiología , Transducción de Señal/inmunología , Animales , Quimiotaxis de Leucocito/genética , Desnervación , Corteza Entorrinal/metabolismo , Regulación de la Expresión Génica/inmunología , Mediadores de Inflamación/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Mutación Puntual , Transducción de Señal/genética , Técnicas Estereotáxicas , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología
16.
J Neuroimmunol ; 203(1): 64-72, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18653241

RESUMEN

We have addressed the role of macrophages in glial response and T cell entry to the CNS after axonal injury, by using intravenous injection of clodronate-loaded mannosylated liposomes, in C57BL6 mice. As expected, clodronate-liposome treatment resulted in depletion of peripheral macrophages which was confirmed by F4/80- and MOMA-1(-) stainings in spleen. Sequential clodronate-liposome treatment 4, 2 and 0 days before axotomy resulted in significant reduction of infiltrating CD45(high) CD11b+ macrophages in the hippocampus at 1, 2 and 3 days post-lesion, measured by flow cytometry. There was a slight delay in the expansion of CD45(dim) CD11+ microglia in clodronate-liposome treated mice, but macrophage depletion had no effect on the percentage of infiltrating T cells in the lesion-reactive hippocampus. Lesion-induced TNFalpha mRNA expression was not affected by macrophage depletion, suggesting that activated glial cells are the primary source of this cytokine in the axonal injury-reactive brain. This identifies a potentially important distinction from inflammatory autoimmune infiltration in EAE, where macrophages are a prominent source of TNFalpha and their depletion prevents parenchymal T cell infiltration and disease.


Asunto(s)
Movimiento Celular/inmunología , Encefalitis/inmunología , Hipocampo/lesiones , Macrófagos/inmunología , Linfocitos T/inmunología , Animales , Axotomía , Conservadores de la Densidad Ósea/farmacología , Comunicación Celular/inmunología , Ácido Clodrónico/farmacología , Encefalitis/patología , Femenino , Citometría de Flujo , Hipocampo/inmunología , Hipocampo/patología , Liposomas , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Microglía/inmunología , Linfocitos T/citología , Factor de Necrosis Tumoral alfa/metabolismo
17.
FASEB J ; 22(8): 2843-52, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18390924

RESUMEN

The generation of new neurons and glia from a precursor stem cell appears to take place in the adult brain. However, new neurons generated in the dentate gyrus decline sharply with age and to an even greater extent in neurodegenerative diseases. Here we raise the question whether peripheral immune mechanisms can generate immunity to such deficits in neuronal repair. We demonstrate that in contrast to primarily innate immunity cytokines, such as interleukin-6 and tumor necrosis factor-alpha, the adaptive immunity cytokine IFN-gamma enhances neurogenesis in the dentate gyrus of adult mice and improves the spatial learning and memory performance of the animals. In older mice, the effect of IFN-gamma is more pronounced in both wild-type mice and mice with Alzheimer's-like disease and is associated with neuroprotection. In addition, IFN-gamma reverses the increase in oligodendrogenesis observed in a mouse model of Alzheimer's disease. We demonstrate that limited amounts of IFN-gamma in the brain shape the neuropoietic milieu to enhance neurogenesis, possibly representing the normal function of the immune system in controlling brain inflammation and repair.


Asunto(s)
Enfermedad de Alzheimer/terapia , Interferón gamma/metabolismo , Regeneración Nerviosa/fisiología , Células Madre Adultas/inmunología , Células Madre Adultas/patología , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/psicología , Precursor de Proteína beta-Amiloide/genética , Animales , Giro Dentado/inmunología , Giro Dentado/metabolismo , Giro Dentado/patología , Modelos Animales de Enfermedad , Hipocampo/inmunología , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Interferón gamma/genética , Aprendizaje por Laberinto , Memoria , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regeneración Nerviosa/inmunología , Neuroinmunomodulación , Neuronas/inmunología , Neuronas/patología , Proteínas Recombinantes/genética , Sinaptofisina/metabolismo
18.
J Immunol ; 178(12): 8175-82, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17548656

RESUMEN

Inflammation of the CNS, which occurs during multiple sclerosis and experimental autoimmune encephalomyelitis, is characterized by increased levels of IFN-gamma, a cytokine not normally expressed in the CNS. To investigate the role of IFN-gamma in CNS, we used intrathecal injection of a replication-defective adenovirus encoding murine IFN-gamma (AdIFNgamma) to IFN-gamma-deficient (GKO) mice. This method resulted in stable, long-lived expression of IFN-gamma that could be detected in cerebrospinal fluid using ELISA and Luminex bead immunoassay. IFN-gamma induced expression in the CNS of message and protein for the chemokines CXCL10 and CCL5, to levels comparable to those seen during experimental autoimmune encephalomyelitis. Other chemokines (CXCL2, CCL2, CCL3) were not induced. Mice lacking the IFN-gammaR showed no response, and a control viral vector did not induce chemokine expression. Chemokine expression was predominantly localized to meningeal and ependymal cells, and was also seen in astrocytes and microglia. IFN-gamma-induced chemokine expression did not lead to inflammation. However, when pertussis toxin was given i.p. to mice infected with the IFN-gamma vector, there was a dramatic increase in the number of T lymphocytes detected in the CNS by flow cytometry. This increase in blood-derived immune cells in the CNS did not occur with pertussis toxin alone, and did not manifest as histologically detectable inflammatory pathology. These results show that IFN-gamma induces a characteristic glial chemokine response that by itself is insufficient to promote inflammation, and that IFN-gamma-induced CNS chemoattractant signals can synergize with a peripheral infectious stimulus to drive T cell entry into the CNS.


Asunto(s)
Movimiento Celular , Sistema Nervioso Central/inmunología , Quimiocinas/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Interferón gamma/fisiología , Linfocitos T/inmunología , Animales , Sistema Nervioso Central/efectos de los fármacos , Quimiocinas/análisis , Interferón gamma/genética , Interferón gamma/farmacología , Ratones , Ratones Noqueados , Neuroglía/inmunología , Toxina del Pertussis/inmunología , Toxina del Pertussis/farmacología , Receptores de Interferón/genética , Linfocitos T/química , Receptor de Interferón gamma
19.
Am J Pathol ; 170(5): 1713-24, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17456776

RESUMEN

We have characterized the cellular response to demyelination/remyelination in the central nervous system using the toxin cuprizone, which causes reproducible demyelination in the corpus callosum. Microglia were distinguished from macrophages by relative CD45 expression (CD45(dim)) using flow cytometry. Their expansion occurred rapidly and substantially outnumbered infiltrating macrophages and T cells throughout the course of cuprizone treatment. We used bromodeoxyuridine incorporation and bone marrow chimeras to show that both proliferation and immigration from blood accounted for increased microglial numbers. Microglia adopted an activated phenotype during demyelination, up-regulating major histocompatibility class I and B7.2/CD86. A subpopulation of CD45(dim-high) microglia that expressed reduced levels of CD11b emerged during demyelination. These microglia expressed CD11c and were potent antigen-presenting cells in vitro. T cells were recruited to the demyelinated corpus callosum but did not appear to be activated. Our study highlights the role of microglia as a heterogeneous population of cells in primary demyelination, with the capacity to present antigen, proliferate, and migrate into demyelinated areas.


Asunto(s)
Cuerpo Calloso/inmunología , Cuerpo Calloso/patología , Enfermedades Desmielinizantes/inmunología , Animales , Proliferación Celular , Cuprizona/toxicidad , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/patología , Femenino , Citometría de Flujo , Inmunohistoquímica , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía , Inhibidores de la Monoaminooxidasa/toxicidad , Linfocitos T/inmunología , Linfocitos T/metabolismo
20.
J Neurosci ; 26(49): 12826-37, 2006 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-17151286

RESUMEN

Reactive gliosis is a prominent feature of neurodegenerative and neuroinflammatory disease in the CNS, yet the stimuli that drive this response are not known. There is growing appreciation that signaling through Toll-like receptors (TLRs), which is key to generating innate responses to infection, may have pathogen-independent roles. We show that TLR2 was selectively upregulated by microglia in the denervated zones of the hippocampus in response to stereotactic transection of axons in the entorhinal cortex. In mice lacking TLR2, there were transient, selective reductions in lesion-induced expression of cytokines and chemokines. Recruitment of T cells, but not macrophages, was delayed in TLR2-deficient mice, as well as in mice lacking TNFR1 (tumor necrosis factor receptor 1). TLR2 deficiency also affected microglial proliferative expansion, whereas all of these events were unaffected in TLR4-mutant mice. Consistent with the fact that responses in knock-out mice had all returned to wild-type levels by 8 d, there was no evidence for effects on neuronal plasticity at 20 d. These results identify a role for TLR2 signaling in the early glial response to brain injury, acting as an innate bridge to neuroinflammation.


Asunto(s)
Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Neuronas/metabolismo , Neuronas/patología , Transducción de Señal/fisiología , Receptor Toll-Like 2/fisiología , Animales , Axones/patología , Lesiones Encefálicas/genética , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Microglía/patología , Transducción de Señal/genética , Receptor Toll-Like 2/biosíntesis , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA