Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(35): e2211310119, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-35994674

RESUMEN

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS). Astrocytes are the most abundant glial cells in the CNS, and their dysfunction contributes to the pathogenesis of MS and its animal model, experimental autoimmune encephalomyelitis (EAE). Recent advances highlight the pivotal role of cellular metabolism in programming immune responses. However, the underlying immunometabolic mechanisms that drive astrocyte pathogenicity remain elusive. Nicotinamide adenine dinucleotide (NAD+) is a vital coenzyme involved in cellular redox reactions and a substrate for NAD+-dependent enzymes. Cellular NAD+ levels are dynamically controlled by synthesis and degradation, and dysregulation of this balance has been associated with inflammation and disease. Here, we demonstrate that cell-autonomous generation of NAD+ via the salvage pathway regulates astrocyte immune function. Inhibition of nicotinamide phosphoribosyltransferase (NAMPT), a key enzyme in the salvage pathway, results in depletion of NAD+, inhibits oxidative phosphorylation, and limits astrocyte inflammatory potential. We identified CD38 as the main NADase up-regulated in reactive mouse and human astrocytes in models of neuroinflammation and MS. Genetic or pharmacological blockade of astrocyte CD38 activity augmented NAD+ levels, suppressed proinflammatory transcriptional reprogramming, impaired chemotactic potential to inflammatory monocytes, and ameliorated EAE. We found that CD38 activity is mediated via calcineurin/NFAT signaling in mouse and human reactive astrocytes. Thus, NAMPT-NAD+-CD38 circuitry in astrocytes controls their ability to meet their energy demands and drives the expression of proinflammatory transcriptional modules, contributing to CNS pathology in EAE and, potentially, MS. Our results identify candidate therapeutic targets in MS.


Asunto(s)
ADP-Ribosil Ciclasa 1 , Astrocitos , Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , NAD , ADP-Ribosil Ciclasa 1/metabolismo , Animales , Astrocitos/inmunología , Astrocitos/metabolismo , Autoinmunidad , Sistema Nervioso Central/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Humanos , Ratones , Esclerosis Múltiple/inmunología , NAD/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo
2.
Brain ; 145(9): 3288-3307, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-35899587

RESUMEN

Malignant brain tumours are the cause of a disproportionate level of morbidity and mortality among cancer patients, an unfortunate statistic that has remained constant for decades. Despite considerable advances in the molecular characterization of these tumours, targeting the cancer cells has yet to produce significant advances in treatment. An alternative strategy is to target cells in the glioblastoma microenvironment, such as tumour-associated astrocytes. Astrocytes control multiple processes in health and disease, ranging from maintaining the brain's metabolic homeostasis, to modulating neuroinflammation. However, their role in glioblastoma pathogenicity is not well understood. Here we report that depletion of reactive astrocytes regresses glioblastoma and prolongs mouse survival. Analysis of the tumour-associated astrocyte translatome revealed astrocytes initiate transcriptional programmes that shape the immune and metabolic compartments in the glioma microenvironment. Specifically, their expression of CCL2 and CSF1 governs the recruitment of tumour-associated macrophages and promotes a pro-tumourigenic macrophage phenotype. Concomitantly, we demonstrate that astrocyte-derived cholesterol is key to glioma cell survival, and that targeting astrocytic cholesterol efflux, via ABCA1, halts tumour progression. In summary, astrocytes control glioblastoma pathogenicity by reprogramming the immunological properties of the tumour microenvironment and supporting the non-oncogenic metabolic dependency of glioblastoma on cholesterol. These findings suggest that targeting astrocyte immunometabolic signalling may be useful in treating this uniformly lethal brain tumour.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Animales , Astrocitos/metabolismo , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Glioma/genética , Ratones , Microambiente Tumoral , Virulencia
3.
Sci Rep ; 11(1): 14644, 2021 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-34282238

RESUMEN

Inhibition of extracellular glutamate (Glu) release decreases proliferation and invasion, induces apoptosis, and inhibits melanoma metastatic abilities. Previous studies have shown that Blood-glutamate scavenging (BGS), a novel treatment approach, has been found to be beneficial in attenuating glioblastoma progression by reducing brain Glu levels. Therefore, in this study we evaluated the ability of BGS treatment to inhibit brain metastatic melanoma progression in-vivo. RET melanoma cells were implanted in C56BL/6J mice to induce brain melanoma tumors followed by treatment with BGS or vehicle administered for fourteen days. Bioluminescent imaging was conducted to evaluate tumor growth, and plasma/CSF Glu levels were monitored throughout. Immunofluorescence staining of Ki67 and 53BP1 was used to analyze tumor cell proliferation and DNA double-strand breaks. In addition, we analyzed CD8, CD68, CD206, p-STAT1 and iNOS expression to evaluate alterations in tumor micro-environment and anti-tumor immune response due to treatment. Our results show that BGS treatment reduces CSF Glu concentration and consequently melanoma growth in-vivo by decreasing tumor cell proliferation and increasing pro-apoptotic signaling in C56BL/6J mice. Furthermore, BGS treatment supported CD8+ cell recruitment and CD68+ macrophage invasion. These findings suggest that BGS can be of potential therapeutic relevance in the treatment of metastatic melanoma.


Asunto(s)
Aspartato Aminotransferasa Citoplasmática/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Ácido Glutámico/metabolismo , Melanoma/tratamiento farmacológico , Ácido Oxaloacético/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Aspartato Aminotransferasa Citoplasmática/farmacología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/secundario , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimioterapia Combinada , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Glioblastoma/secundario , Humanos , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Terapia Molecular Dirigida/métodos , Ácido Oxaloacético/farmacología , Proteínas Recombinantes/administración & dosificación , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
4.
Cell ; 179(7): 1483-1498.e22, 2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31813625

RESUMEN

Metabolism has been shown to control peripheral immunity, but little is known about its role in central nervous system (CNS) inflammation. Through a combination of proteomic, metabolomic, transcriptomic, and perturbation studies, we found that sphingolipid metabolism in astrocytes triggers the interaction of the C2 domain in cytosolic phospholipase A2 (cPLA2) with the CARD domain in mitochondrial antiviral signaling protein (MAVS), boosting NF-κB-driven transcriptional programs that promote CNS inflammation in experimental autoimmune encephalomyelitis (EAE) and, potentially, multiple sclerosis. cPLA2 recruitment to MAVS also disrupts MAVS-hexokinase 2 (HK2) interactions, decreasing HK enzymatic activity and the production of lactate involved in the metabolic support of neurons. Miglustat, a drug used to treat Gaucher and Niemann-Pick disease, suppresses astrocyte pathogenic activities and ameliorates EAE. Collectively, these findings define a novel immunometabolic mechanism that drives pro-inflammatory astrocyte activities, outlines a new role for MAVS in CNS inflammation, and identifies candidate targets for therapeutic intervention.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Astrocitos/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Fosfolipasas A2 Secretoras/metabolismo , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/farmacología , 1-Desoxinojirimicina/uso terapéutico , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Astrocitos/efectos de los fármacos , Astrocitos/patología , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , Hexoquinasa/metabolismo , Humanos , Ácido Láctico/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Fosfolipasas A2 Secretoras/genética
6.
Nat Neurosci ; 22(5): 729-740, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30962630

RESUMEN

Tumor-associated macrophages (TAMs) play an important role in the immune response to cancer, but the mechanisms by which the tumor microenvironment controls TAMs and T cell immunity are not completely understood. Here we report that kynurenine produced by glioblastoma cells activates aryl hydrocarbon receptor (AHR) in TAMs to modulate their function and T cell immunity. AHR promotes CCR2 expression, driving TAM recruitment in response to CCL2. AHR also drives the expression of KLF4 and suppresses NF-κB activation in TAMs. Finally, AHR drives the expression of the ectonucleotidase CD39 in TAMs, which promotes CD8+ T cell dysfunction by producing adenosine in cooperation with CD73. In humans, the expression of AHR and CD39 was highest in grade 4 glioma, and high AHR expression was associated with poor prognosis. In summary, AHR and CD39 expressed in TAMs participate in the regulation of the immune response in glioblastoma and constitute potential targets for immunotherapy.


Asunto(s)
Antígenos CD/metabolismo , Apirasa/metabolismo , Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Quinurenina/metabolismo , Macrófagos/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Linfocitos T/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Glioblastoma/metabolismo , Humanos , Factor 4 Similar a Kruppel , Receptores de Lipopolisacáridos/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/metabolismo , Factor de Transcripción STAT1 , Factor de Transcripción STAT3/metabolismo , Linfocitos T/inmunología , Microambiente Tumoral
7.
Nat Methods ; 16(4): 327-332, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30886410

RESUMEN

Single-cell RNA sequencing (scRNA-seq) is a rich resource of cellular heterogeneity, opening new avenues in the study of complex tissues. We introduce Cell Population Mapping (CPM), a deconvolution algorithm in which reference scRNA-seq profiles are leveraged to infer the composition of cell types and states from bulk transcriptome data ('scBio' CRAN R-package). Analysis of individual variations in lungs of influenza-virus-infected mice reveals that the relationship between cell abundance and clinical symptoms is a cell-state-specific property that varies gradually along the continuum of cell-activation states. The gradual change is confirmed in subsequent experiments and is further explained by a mathematical model in which clinical outcomes relate to cell-state dynamics along the activation process. Our results demonstrate the power of CPM in reconstructing the continuous spectrum of cell states within heterogeneous tissues.


Asunto(s)
Biología Computacional , Genómica , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Algoritmos , Animales , Separación Celular , Femenino , Fibroblastos/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Genoma Humano , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Pulmón/virología , Cadenas de Markov , Ratones , Ratones Endogámicos C57BL , Orthomyxoviridae , Fagocitos/metabolismo , Valores de Referencia , Programas Informáticos , Transcriptoma
8.
PLoS Biol ; 17(3): e2006859, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30921319

RESUMEN

Brain metastases are prevalent in various types of cancer and are often terminal, given the low efficacy of available therapies. Therefore, preventing them is of utmost clinical relevance, and prophylactic treatments are perhaps the most efficient strategy. Here, we show that systemic prophylactic administration of a toll-like receptor (TLR) 9 agonist, CpG-C, is effective against brain metastases. Acute and chronic systemic administration of CpG-C reduced tumor cell seeding and growth in the brain in three tumor models in mice, including metastasis of human and mouse lung cancer, and spontaneous melanoma-derived brain metastasis. Studying mechanisms underlying the therapeutic effects of CpG-C, we found that in the brain, unlike in the periphery, natural killer (NK) cells and monocytes are not involved in controlling metastasis. Next, we demonstrated that the systemically administered CpG-C is taken up by endothelial cells, astrocytes, and microglia, without affecting blood-brain barrier (BBB) integrity and tumor brain extravasation. In vitro assays pointed to microglia, but not astrocytes, as mediators of CpG- C effects through increased tumor killing and phagocytosis, mediated by direct microglia-tumor contact. In vivo, CpG-C-activated microglia displayed elevated mRNA expression levels of apoptosis-inducing and phagocytosis-related genes. Intravital imaging showed that CpG-C-activated microglia cells contact, kill, and phagocytize tumor cells in the early stages of tumor brain invasion more than nonactivated microglia. Blocking in vivo activation of microglia with minocycline, and depletion of microglia with a colony-stimulating factor 1 inhibitor, indicated that microglia mediate the antitumor effects of CpG-C. Overall, the results suggest prophylactic CpG-C treatment as a new intervention against brain metastasis, through an essential activation of microglia.


Asunto(s)
Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/metabolismo , Microglía/metabolismo , Microglía/patología , Oligodesoxirribonucleótidos/uso terapéutico , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Factores Estimulantes de Colonias/antagonistas & inhibidores , Factores Estimulantes de Colonias/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/complicaciones , Neoplasias Pulmonares/metabolismo , Masculino , Melanoma/complicaciones , Melanoma/metabolismo , Ratones , Minociclina/metabolismo , Fagocitosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
9.
Nat Commun ; 9(1): 4578, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30385785

RESUMEN

Previous studies have reported that microglia depletion leads to impairment of synapse formation and these cells rapidly repopulate from CNS progenitors. However, the impact of microglia depletion and repopulation in the long-term state of the CNS environment has not been characterized. Here, we report that acute and synchronous microglia depletion and subsequent repopulation induces gray matter microgliosis, neuronal death in the somatosensory cortex and ataxia-like behavior. We find a type 1 interferon inflammatory signature in degenerating somatosensory cortex from microglia-depleted mice. Transcriptomic and mass cytometry analysis of repopulated microglia demonstrates an interferon regulatory factor 7-driven activation state. Minocycline and anti-IFNAR1 antibody treatment attenuate the CNS type 1 interferon-driven inflammation, restore microglia homeostasis and reduce ataxic behavior. Neither microglia depletion nor repopulation impact neuropathology or T-cell responses during experimental autoimmune encephalomyelitis. Together, we found that acute microglia ablation induces a type 1 interferon activation state of gray matter microglia associated with acute neurodegeneration.


Asunto(s)
Muerte Celular/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Sustancia Gris/inmunología , Interferón Tipo I/inmunología , Microglía/inmunología , Neuronas/inmunología , Corteza Somatosensorial/inmunología , Animales , Antibacterianos/farmacología , Ataxia/inmunología , Ataxia/patología , Encefalomielitis Autoinmune Experimental/patología , Citometría de Flujo , Perfilación de la Expresión Génica , Sustancia Gris/patología , Homeostasis , Inmunohistoquímica , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/inmunología , Ratones , Microscopía Confocal , Minociclina/farmacología , Neuronas/patología , Receptor de Interferón alfa y beta/antagonistas & inhibidores , Prueba de Desempeño de Rotación con Aceleración Constante , Corteza Somatosensorial/patología
10.
Cell Syst ; 6(6): 679-691.e4, 2018 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-29886109

RESUMEN

The influenza virus is a major cause of morbidity and mortality worldwide. Yet, both the impact of intracellular viral replication and the variation in host response across different cell types remain uncharacterized. Here we used single-cell RNA sequencing to investigate the heterogeneity in the response of lung tissue cells to in vivo influenza infection. Analysis of viral and host transcriptomes in the same single cell enabled us to resolve the cellular heterogeneity of bystander (exposed but uninfected) as compared with infected cells. We reveal that all major immune and non-immune cell types manifest substantial fractions of infected cells, albeit at low viral transcriptome loads relative to epithelial cells. We show that all cell types respond primarily with a robust generic transcriptional response, and we demonstrate novel markers specific for influenza-infected as opposed to bystander cells. These findings open new avenues for targeted therapy aimed exclusively at infected cells.


Asunto(s)
Interacciones Huésped-Patógeno/genética , Gripe Humana/genética , Orthomyxoviridae/genética , Animales , Secuencia de Bases/genética , Línea Celular , Células Epiteliales/inmunología , Femenino , Perfilación de la Expresión Génica/métodos , Interacciones Huésped-Patógeno/inmunología , Humanos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/genética , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Transcriptoma/genética , Replicación Viral
11.
Science ; 353(6299): 603-8, 2016 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-27493188

RESUMEN

Mutations in the optineurin (OPTN) gene have been implicated in both familial and sporadic amyotrophic lateral sclerosis (ALS). However, the role of this protein in the central nervous system (CNS) and how it may contribute to ALS pathology are unclear. Here, we found that optineurin actively suppressed receptor-interacting kinase 1 (RIPK1)-dependent signaling by regulating its turnover. Loss of OPTN led to progressive dysmyelination and axonal degeneration through engagement of necroptotic machinery in the CNS, including RIPK1, RIPK3, and mixed lineage kinase domain-like protein (MLKL). Furthermore, RIPK1- and RIPK3-mediated axonal pathology was commonly observed in SOD1(G93A) transgenic mice and pathological samples from human ALS patients. Thus, RIPK1 and RIPK3 play a critical role in mediating progressive axonal degeneration. Furthermore, inhibiting RIPK1 kinase may provide an axonal protective strategy for the treatment of ALS and other human degenerative diseases characterized by axonal degeneration.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Apoptosis , Axones/patología , Degeneración Nerviosa/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/fisiología , Factor de Transcripción TFIIIA/metabolismo , Esclerosis Amiotrófica Lateral/genética , Animales , Apoptosis/genética , Proteínas de Ciclo Celular , Humanos , Inflamación/genética , Inflamación/patología , Proteínas de Transporte de Membrana , Ratones , Ratones Transgénicos , Necrosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Médula Espinal/patología , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Supresión Genética , Factor de Transcripción TFIIIA/genética
12.
Brain ; 139(Pt 7): 1939-57, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27246324

RESUMEN

SEE WINGER AND ZAMVIL DOI101093/BRAIN/AWW121 FOR A SCIENTIFIC COMMENTARY ON THIS ARTICLE: The innate immune system plays a central role in the chronic central nervous system inflammation that drives neurological disability in progressive forms of multiple sclerosis, for which there are no effective treatments. The mucosal immune system is a unique tolerogenic organ that provides a physiological approach for the induction of regulatory T cells. Here we report that nasal administration of CD3-specific antibody ameliorates disease in a progressive animal model of multiple sclerosis. This effect is IL-10-dependent and is mediated by the induction of regulatory T cells that share a similar transcriptional profile to Tr1 regulatory cells and that suppress the astrocyte inflammatory transcriptional program. Treatment results in an attenuated inflammatory milieu in the central nervous system, decreased microglia activation, reduced recruitment of peripheral monocytes, stabilization of the blood-brain barrier and less neurodegeneration. These findings suggest a new therapeutic approach for the treatment of progressive forms of multiple sclerosis and potentially other types of chronic central nervous system inflammation.


Asunto(s)
Astrocitos/inmunología , Complejo CD3/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Factores Inmunológicos/farmacología , Interleucina-10/inmunología , Muromonab-CD3/farmacología , Linfocitos T Reguladores/inmunología , Administración Intranasal , Animales , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , Factores Inmunológicos/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Muromonab-CD3/administración & dosificación , Neumonía Neumocócica/inmunología
13.
Nat Med ; 22(6): 586-97, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27158906

RESUMEN

Astrocytes have important roles in the central nervous system (CNS) during health and disease. Through genome-wide analyses we detected a transcriptional response to type I interferons (IFN-Is) in astrocytes during experimental CNS autoimmunity and also in CNS lesions from patients with multiple sclerosis (MS). IFN-I signaling in astrocytes reduces inflammation and experimental autoimmune encephalomyelitis (EAE) disease scores via the ligand-activated transcription factor aryl hydrocarbon receptor (AHR) and the suppressor of cytokine signaling 2 (SOCS2). The anti-inflammatory effects of nasally administered interferon (IFN)-ß are partly mediated by AHR. Dietary tryptophan is metabolized by the gut microbiota into AHR agonists that have an effect on astrocytes to limit CNS inflammation. EAE scores were increased following ampicillin treatment during the recovery phase, and CNS inflammation was reduced in antibiotic-treated mice by supplementation with the tryptophan metabolites indole, indoxyl-3-sulfate, indole-3-propionic acid and indole-3-aldehyde, or the bacterial enzyme tryptophanase. In individuals with MS, the circulating levels of AHR agonists were decreased. These findings suggest that IFN-Is produced in the CNS function in combination with metabolites derived from dietary tryptophan by the gut flora to activate AHR signaling in astrocytes and suppress CNS inflammation.


Asunto(s)
Astrocitos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Microbioma Gastrointestinal , Interferón Tipo I/inmunología , Esclerosis Múltiple/inmunología , Receptores de Hidrocarburo de Aril/inmunología , Linfocitos T/inmunología , Triptófano/metabolismo , Animales , Estudios de Casos y Controles , Proliferación Celular , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Quimiocina CCL2/metabolismo , Inmunoprecipitación de Cromatina , Cromatografía Líquida de Alta Presión , Encefalomielitis Autoinmune Experimental/metabolismo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Immunoblotting , Indicán/orina , Indoles/metabolismo , Inflamación , Interferón beta/farmacología , Limosilactobacillus reuteri , Ratones , Ratones Noqueados , Esclerosis Múltiple/metabolismo , Proteínas de Resistencia a Mixovirus/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Imagen Óptica , Reacción en Cadena de la Polimerasa , Receptor de Interferón alfa y beta/genética , Receptores de Hidrocarburo de Aril/metabolismo , Factor de Transcripción STAT1/metabolismo , Serotonina , Proteínas Supresoras de la Señalización de Citocinas , Triptofanasa/metabolismo
14.
Nat Med ; 20(10): 1147-56, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25216636

RESUMEN

Astrocytes have complex roles in health and disease, thus it is important to study the pathways that regulate their function. Here we report that lactosylceramide (LacCer) synthesized by ß-1,4-galactosyltransferase 6 (B4GALT6) is upregulated in the central nervous system (CNS) of mice during chronic experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (MS). LacCer acts in an autocrine manner to control astrocyte transcriptional programs that promote neurodegeneration. In addition, LacCer in astrocytes controls the recruitment and activation of microglia and CNS-infiltrating monocytes in a non-cell autonomous manner by regulating production of the chemokine CCL2 and granulocyte-macrophage colony-stimulating factor (GM-CSF), respectively. We also detected high B4GALT6 gene expression and LacCer concentrations in CNS MS lesions. Inhibition of LacCer synthesis in mice suppressed local CNS innate immunity and neurodegeneration in EAE and interfered with the activation of human astrocytes in vitro. Thus, B4GALT6 regulates astrocyte activation and is a potential therapeutic target for MS and other neuroinflammatory disorders.


Asunto(s)
Astrocitos/inmunología , Astrocitos/metabolismo , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Galactosiltransferasas/metabolismo , Glucolípidos/metabolismo , Animales , Antígenos CD/metabolismo , Sistema Nervioso Central/patología , Quimiocina CCL2/genética , Encefalomielitis Autoinmune Experimental/genética , Femenino , Galactosiltransferasas/genética , Técnicas de Silenciamiento del Gen , Proteína Ácida Fibrilar de la Glía , Humanos , Inmunidad Innata , Lactosilceramidos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Degeneración Nerviosa/genética , Degeneración Nerviosa/inmunología , Degeneración Nerviosa/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Regulación hacia Arriba
15.
Nat Immunol ; 14(10): 1054-63, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23995234

RESUMEN

Dendritic cells (DCs) control the balance between effector T cells and regulatory T cells in vivo. Hence, the study of DCs might identify mechanisms of disease pathogenesis and guide new therapeutic approaches for disorders mediated by the immune system. We found that interleukin 27 (IL-27) signaling in mouse DCs limited the generation of effector cells of the TH1 and TH17 subsets of helper T cells and the development of experimental autoimmune encephalomyelitis (EAE). The effects of IL-27 were mediated at least in part through induction of the immunoregulatory molecule CD39 in DCs. IL-27-induced CD39 decreased the extracellular concentration of ATP and downregulated nucleotide-dependent activation of the NLRP3 inflammasome. Finally, therapeutic vaccination with IL-27-conditioned DCs suppressed established relapsing-remitting EAE. Thus, IL-27 signaling in DCs limited pathogenic T cell responses and the development of autoimmunity.


Asunto(s)
Antígenos CD/genética , Apirasa/genética , Autoinmunidad , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Interleucina-17/farmacología , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Animales , Anticuerpos/inmunología , Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/inmunología , Antígenos CD/metabolismo , Apirasa/metabolismo , Autoanticuerpos/inmunología , Autoinmunidad/efectos de los fármacos , Proteínas Portadoras/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Citocinas/biosíntesis , Células Dendríticas/metabolismo , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Tolerancia Inmunológica/inmunología , Ratones , Ratones Noqueados , Vaina de Mielina/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo , Receptores de Interleucina , Transducción de Señal , Subgrupos de Linfocitos T/citología , Transcripción Genética/efectos de los fármacos
16.
Glia ; 61(6): 985-1002, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23536313

RESUMEN

Transforming growth factor ß1 (TGF-ß1) is a pleiotropic cytokine expressed throughout the CNS. Previous studies demonstrated that TGF-ß1 contributes to maintain neuronal survival, but mechanistically this effect is not well understood. We generated a CNS-specific TGF-ß1-deficient mouse model to investigate the functional consequences of TGF-ß1-deficiency in the adult mouse brain. We found that depletion of TGF-ß1 in the CNS resulted in a loss of the astrocyte glutamate transporter (GluT) proteins GLT-1 (EAAT2) and GLAST (EAAT1) and decreased glutamate uptake in the mouse hippocampus. Treatment with TGF-ß1 induced the expression of GLAST and GLT-1 in cultured astrocytes and enhanced astroglial glutamate uptake. Similar to GLT-1-deficient mice, CNS-TGF-ß1-deficient mice had reduced brain weight and neuronal loss in the CA1 hippocampal region. CNS-TGF-ß1-deficient mice showed GluN2B-dependent aberrant synaptic plasticity in the CA1 area of the hippocampus similar to the glutamate transport inhibitor DL-TBOA and these mice were highly sensitive to excitotoxic injury. In addition, hippocampal neurons from TGF-ß1-deficient mice had elevated GluN2B-mediated calcium signals in response to extrasynaptic glutamate receptor stimulation, whereas cells treated with TGF-ß1 exhibited reduced GluN2B-mediated calcium signals. In summary, our study demonstrates a previously unrecognized function of TGF-ß1 in the CNS to control extracellular glutamate homeostasis and GluN2B-mediated calcium responses in the mouse hippocampus.


Asunto(s)
Calcio/metabolismo , Ácido Glutámico/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Apoptosis/fisiología , Espinas Dendríticas/metabolismo , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 1 de Aminoácidos Excitadores/metabolismo , Transportador 2 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/metabolismo , Hipocampo/metabolismo , Ratones , Ratones Noqueados , Transmisión Sináptica/fisiología , Factor de Crecimiento Transformador beta1/genética
17.
Neuro Oncol ; 14(8): 1037-49, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22700727

RESUMEN

Gliomas are the most frequent primary tumors of the brain, and for highly malignant gliomas there is no successful treatment. The tumor microenvironment contains large numbers of infiltrating microglia and macrophages (MM). There is increasing evidence that the tumor-associated MM support glioma expansion. CD38 is a multifunctional ectoenzyme that uses nicotinamide adenine dinucleotide as a substrate to generate second messengers. Previously we showed that CD38 deficiency modulates microglial "activation" and impaired recovery from head trauma by a microglia-associated mechanism. In view of the supportive role of MM in glioma progression and the role of CD38 in microglia activation, we hypothesize that deficiency of CD38 in the tumor microenvironment would inhibit glioma progression. Using the syngeneic GL261 model of glioma progression in wild-type and CD38 null mice, we show here that CD38 deficiency significantly attenuates glioma expansion and prolongs the life span of the glioma-bearing mice. The CD38 deficiency effect was associated with increased cell death and decreased metalloproteinase-12 expression in the tumor mass, as well as modulation of the tumor-induced MM properties, as indicated by a reduction in the expression of the MM marker F4/80 and matrix metalloproteinases. Our results thus suggest that CD38 participates in the tumor-supporting action of MM and that targeting CD38 might be a potential therapeutic approach for glioma treatment.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Neoplasias Encefálicas/patología , Glioma/patología , Macrófagos/patología , Glicoproteínas de Membrana/metabolismo , Microglía/patología , Microambiente Tumoral/inmunología , ADP-Ribosil Ciclasa 1/deficiencia , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/metabolismo , Separación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Citometría de Flujo , Glioma/inmunología , Glioma/metabolismo , Immunoblotting , Inmunohistoquímica , Macrófagos/inmunología , Masculino , Glicoproteínas de Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Immunol Rev ; 248(1): 170-87, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22725961

RESUMEN

Demyelinating diseases such as multiple sclerosis are chronic inflammatory autoimmune diseases with a heterogeneous clinical presentation and course. Both the adaptive and the innate immune systems have been suggested to contribute to their pathogenesis and recovery. In this review, we discuss the role of the innate immune system in mediating demyelinating diseases. In particular, we provide an overview of the anti-inflammatory or pro-inflammatory functions of dendritic cells, mast cells, natural killer (NK) cells, NK-T cells, γδ T cells, microglial cells, and astrocytes. We emphasize the interaction of astroctyes with the immune system and how this interaction relates to the demyelinating pathologies. Given the pivotal role of the innate immune system, it is possible that targeting these cells may provide an effective therapeutic approach for demyelinating diseases.


Asunto(s)
Enfermedades Desmielinizantes/inmunología , Inmunidad Innata , Inmunidad Adaptativa , Animales , Astrocitos/inmunología , Astrocitos/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Enfermedades Desmielinizantes/metabolismo , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Mastocitos/inmunología , Mastocitos/metabolismo , Sistema Mononuclear Fagocítico/inmunología , Sistema Mononuclear Fagocítico/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
19.
Glia ; 59(3): 397-412, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21264947

RESUMEN

Apoptosis is a controlled cell-death process mediated inter alia by proteins of the Bcl-2 family. Some proteins previously shown to promote the apoptotic process were found to have nonapoptotic functions as well. Microglia, the resident immune cells of the central nervous system, respond to brain derangements by becoming activated to contend with the brain damage. Activated microglia can also undergo activation-induced cell death. Previous studies have addressed the role of core apoptotic proteins in the death process, but whether these proteins also play a role or not in the activation process is not been reported. Here we explore the effect of the BH3-only protein Bid on the immunological features of microglia and macrophages. Our results showed that Bid regulates both the phagocytotic activities and the inflammatory profiles of these cells. Deficiency of Bid attenuated the phagocytotic activity of primary microglia and peritoneal macrophages. It also changed the expression profile of distinct inflammation-related genes in lipopolysaccharide-activated microglia and peritoneal macrophages in vitro and in an in vivo sepsis-like paradigm. Notably, similar changes followed downregulation of Bid in the N9 microglial cell line. Cell death could not be detected in any of the systems examined. Our findings demonstrate that Bid can regulate the immunological profiles of activated microglial and macrophages, via a novel nonapoptotic activity. In view of the critical role of these cells in various pathologies, including acute and chronic brain insults, our findings suggest that impairments in Bid expression may contribute to these pathologies also via a nonapoptotic activity.


Asunto(s)
Proteína Proapoptótica que Interacciona Mediante Dominios BH3/fisiología , Macrófagos/inmunología , Microglía/inmunología , Degeneración Nerviosa/inmunología , Animales , Apoptosis/inmunología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/antagonistas & inhibidores , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/genética , Línea Celular , Células Cultivadas , Femenino , Células HEK293 , Humanos , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Lipopolisacáridos/toxicidad , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/patología , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Fagocitosis/inmunología , Transducción de Señal/inmunología
20.
J Neurotrauma ; 26(9): 1521-33, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19257806

RESUMEN

Traumatic brain injury (TBI) is a major cause of death and disability worldwide. It causes progressive tissue atrophy and consequent neurological dysfunctions. TBI is accompanied by neuroinflammation, a process mediated largely by microglia. CD38 is an ectoenzyme that promotes transmembrane signaling via the synthesis of potent calcium mobilizing agents or via its receptor activity. CD38 is expressed in the brain in various cell types including microglia. In previous studies, we showed that CD38 regulates microglial activation and response to chemokines. In view of the important role of neuroinflammation in TBI and the effects of CD38 on microglial responses, the present study examines the role of CD38 in the recovery of mice from closed head injury (CHI), a model of focal TBI. For this purpose, CD38-deficient and wild-type (WT) mice were subjected to a similar severity of CHI and the effect of the injury on neurobehavioral and cognitive functions was assessed by the Neurological Severity Score (NSS) and the Object Recognition Test, at various time points post-injury. The results show that recovery after CHI (as indicated by the NSS) was significantly lower in CD38-deficient mice than in WT mice and that the object recognition performance after injury was significantly impaired in injured CD38-deficient mice than in WT mice. In addition, we also observed that the amount of activated microglia/macrophages at the injury site was significantly lower in CD38-deficient mice compared with WT mice. Taken together, our findings indicate that CD38 plays a beneficial role in the recovery of mice from CHI and that this effect is mediated, at least in part, via the effect of CD38 on microglia responses.


Asunto(s)
ADP-Ribosil Ciclasa 1/fisiología , Lesiones Encefálicas/genética , Lesiones Encefálicas/patología , ADP-Ribosil Ciclasa 1/genética , Animales , Astrocitos/patología , Conducta Animal , Lesiones Encefálicas/psicología , Traumatismos Cerrados de la Cabeza/genética , Traumatismos Cerrados de la Cabeza/patología , Traumatismos Cerrados de la Cabeza/psicología , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Macrófagos/patología , Memoria/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Microglía/patología , Neuronas/patología , Reconocimiento en Psicología/fisiología , Recuperación de la Función , Índices de Gravedad del Trauma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...