Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep Med ; 5(5): 101546, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38703766

RESUMEN

Mutations in SOD1 cause amyotrophic lateral sclerosis (ALS), a neurodegenerative disease characterized by motor neuron (MN) loss. We previously discovered that macrophage migration inhibitory factor (MIF), whose levels are extremely low in spinal MNs, inhibits mutant SOD1 misfolding and toxicity. In this study, we show that a single peripheral injection of adeno-associated virus (AAV) delivering MIF into adult SOD1G37R mice significantly improves their motor function, delays disease progression, and extends survival. Moreover, MIF treatment reduces neuroinflammation and misfolded SOD1 accumulation, rescues MNs, and corrects dysregulated pathways as observed by proteomics and transcriptomics. Furthermore, we reveal low MIF levels in human induced pluripotent stem cell-derived MNs from familial ALS patients with different genetic mutations, as well as in post mortem tissues of sporadic ALS patients. Our findings indicate that peripheral MIF administration may provide a potential therapeutic mechanism for modulating misfolded SOD1 in vivo and disease outcome in ALS patients.


Asunto(s)
Esclerosis Amiotrófica Lateral , Factores Inhibidores de la Migración de Macrófagos , Neuronas Motoras , Superóxido Dismutasa-1 , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Factores Inhibidores de la Migración de Macrófagos/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/terapia , Esclerosis Amiotrófica Lateral/patología , Animales , Humanos , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Ratones , Células Madre Pluripotentes Inducidas/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Oxidorreductasas Intramoleculares/genética , Ratones Transgénicos , Dependovirus/genética , Modelos Animales de Enfermedad , Masculino , Mutación/genética , Femenino , Pliegue de Proteína
2.
Mol Psychiatry ; 25(5): 1050-1067, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31772304

RESUMEN

Viral infection during pregnancy is often associated with neuropsychiatric conditions. In mice, exposure of pregnant dams to the viral mimetic poly(I:C), serves as a model that simulates such pathology in the offspring, through a process known as Maternal Immune Activation (MIA). To investigate the mechanism of such effect, we hypothesized that maternal upregulation of Type-I interferon (IFN-I), as part of the dam's antiviral response, might contribute to the damage imposed on the offspring. Using mRNA sequencing and flow cytometry analyses we found that poly(I:C) treatment during pregnancy caused reduced expression of genes related to proliferation and cell cycle in the offspring's microglia relative to controls. This was found to be associated with an IFN-I signature in the embryonic yolk sac, the origin of microglia in development. Neutralizing IFN-I signaling in dams attenuated the effect of MIA on the newborn's microglia, while systemic maternal administration of IFNß was sufficient to mimic the effect of poly(I:C), and led to increased vulnerability of offspring's microglia to subsequent stress. Furthermore, maternal elevation of IFNß resulted in behavioral manifestations reminiscent of neuropsychiatric disorders. In addition, by adopting a "two-hit" experimental paradigm, we show a higher sensitivity of the offspring to postnatal stress subsequent to the maternal IFNß elevation, demonstrated by behavioral irregularities. Our results suggest that maternal upregulation of IFN-I, in response to MIA, interferes with the offspring's programmed microglial developmental cascade, increases their susceptibility to postnatal stress, and leads to behavioral abnormalities.


Asunto(s)
Conducta Animal , Interferón Tipo I/inmunología , Microglía/inmunología , Madres/psicología , Efectos Tardíos de la Exposición Prenatal/inmunología , Estrés Psicológico/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Exposición Materna , Ratones , Ratones Endogámicos C57BL , Embarazo
3.
Nat Commun ; 8(1): 717, 2017 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-28959042

RESUMEN

During ageing, microglia acquire a phenotype that may negatively affect brain function. Here we show that ageing microglial phenotype is largely imposed by interferon type I (IFN-I) chronically present in aged brain milieu. Overexpression of IFN-ß in the CNS of adult wild-type mice, but not of mice lacking IFN-I receptor on their microglia, induces an ageing-like transcriptional microglial signature, and impairs cognitive performance. Furthermore, we demonstrate that age-related IFN-I milieu downregulates microglial myocyte-specific enhancer factor 2C (Mef2C). Immune challenge in mice lacking Mef2C in microglia results in an exaggerated microglial response and has an adverse effect on mice behaviour. Overall, our data indicate that the chronic presence of IFN-I in the brain microenvironment, which negatively affects cognitive function, is mediated via modulation of microglial activity. These findings may shed new light on other neurological conditions characterized by elevated IFN-I signalling in the brain.Microglia cells in the brain regulate immune responses, but in ageing can negatively affect brain function. Here the authors show that the chronic presence of type I interferon in aged mouse brain impedes cognitive ability by altering microglia transcriptome and limiting Mef2C, a microglia 'off' signal.


Asunto(s)
Envejecimiento/inmunología , Encéfalo/inmunología , Interferón beta/inmunología , Factores de Transcripción MEF2/inmunología , Microglía/inmunología , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/fisiopatología , Humanos , Interferón beta/genética , Factores de Transcripción MEF2/genética , Masculino , Ratones , Ratones Endogámicos C57BL
4.
Nat Med ; 22(2): 135-7, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26779813

RESUMEN

Systemic immune suppression may curtail the ability to mount the protective, cell-mediated immune responses that are needed for brain repair. By using mouse models of Alzheimer's disease (AD), we show that immune checkpoint blockade directed against the programmed death-1 (PD-1) pathway evokes an interferon (IFN)-γ-dependent systemic immune response, which is followed by the recruitment of monocyte-derived macrophages to the brain. When induced in mice with established pathology, this immunological response leads to clearance of cerebral amyloid-ß (Aß) plaques and improved cognitive performance. Repeated treatment sessions were required to maintain a long-lasting beneficial effect on disease pathology. These findings suggest that immune checkpoints may be targeted therapeutically in AD.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Anticuerpos/farmacología , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Macrófagos/efectos de los fármacos , Memoria/efectos de los fármacos , Placa Amiloide/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/efectos de los fármacos , Péptidos beta-Amiloides/inmunología , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/inmunología , Encéfalo/patología , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Citometría de Flujo , Humanos , Inmunohistoquímica , Interferón gamma/efectos de los fármacos , Interferón gamma/inmunología , Macrófagos/inmunología , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Placa Amiloide/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Análisis de Secuencia de ARN , Transducción de Señal/efectos de los fármacos
5.
Nat Commun ; 6: 7967, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26284939

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder in which chronic neuroinflammation contributes to disease escalation. Nevertheless, while immunosuppressive drugs have repeatedly failed in treating this disease, recruitment of myeloid cells to the CNS was shown to play a reparative role in animal models. Here we show, using the 5XFAD AD mouse model, that transient depletion of Foxp3(+) regulatory T cells (Tregs), or pharmacological inhibition of their activity, is followed by amyloid-ß plaque clearance, mitigation of the neuroinflammatory response and reversal of cognitive decline. We further show that transient Treg depletion affects the brain's choroid plexus, a selective gateway for immune cell trafficking to the CNS, and is associated with subsequent recruitment of immunoregulatory cells, including monocyte-derived macrophages and Tregs, to cerebral sites of plaque pathology. Our findings suggest targeting Treg-mediated systemic immunosuppression for treating AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Factores de Transcripción Forkhead/metabolismo , Linfocitos T Reguladores/metabolismo , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Giro Dentado/patología , Giro Dentado/fisiología , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica/fisiología , Acetato de Glatiramer/farmacología , Tolerancia Inmunológica , Inmunomodulación , Ratones , Ratones Transgénicos , ARN/genética , ARN/metabolismo
6.
EMBO J ; 34(13): 1816-28, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-25940071

RESUMEN

Chronic neuroinflammation is evident in brain aging and neurodegenerative disorders and is often associated with excessive nitric oxide (NO) production within the central nervous system (CNS). Under such conditions, increased NO levels are observed at the choroid plexus (CP), an epithelial layer that forms the blood-cerebrospinal fluid barrier (BCSFB) and serves as a selective gateway for leukocyte entry to the CNS in homeostasis and following injury. Here, we hypothesized that elevated cerebral NO levels interfere with CP gateway activity. We found that induction of leukocyte trafficking determinants by the CP and sequential leukocyte entry to the CSF are dependent on the CP epithelial NFκB/p65 signaling pathway, which was inhibited upon exposure to NO. Examining the CP in 5XFAD transgenic mouse model of Alzheimer's disease (AD-Tg) revealed impaired ability to mount an NFκB/p65-dependent response. Systemic administration of an NO scavenger in AD-Tg mice alleviated NFκB/p65 suppression at the CP and augmented its gateway activity. Together, our findings identify cerebral NO as a negative regulator of CP gateway activity for immune cell trafficking to the CNS.


Asunto(s)
Encéfalo/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Plexo Coroideo/efectos de los fármacos , Leucocitos/efectos de los fármacos , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico/farmacología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/inmunología , Células Cultivadas , Plexo Coroideo/inmunología , Plexo Coroideo/metabolismo , Modelos Animales de Enfermedad , Femenino , Leucocitos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
7.
Science ; 346(6205): 89-93, 2014 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-25147279

RESUMEN

Aging-associated cognitive decline is affected by factors produced inside and outside the brain. By using multiorgan genome-wide analysis of aged mice, we found that the choroid plexus, an interface between the brain and the circulation, shows a type I interferon (IFN-I)-dependent gene expression profile that was also found in aged human brains. In aged mice, this response was induced by brain-derived signals, present in the cerebrospinal fluid. Blocking IFN-I signaling within the aged brain partially restored cognitive function and hippocampal neurogenesis and reestablished IFN-II-dependent choroid plexus activity, which is lost in aging. Our data identify a chronic aging-induced IFN-I signature, often associated with antiviral response, at the brain's choroid plexus and demonstrate its negative influence on brain function, thereby suggesting a target for ameliorating cognitive decline in aging.


Asunto(s)
Envejecimiento/patología , Encéfalo/fisiología , Plexo Coroideo/metabolismo , Cognición , Regulación de la Expresión Génica , Factores Reguladores del Interferón/genética , Interferón Tipo I/fisiología , Envejecimiento/genética , Animales , Hipocampo/citología , Ratones , Ratones Transgénicos , Neurogénesis , Receptores de Interferón/genética , Receptor de Interferón gamma
8.
Brain ; 136(Pt 11): 3427-40, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24088808

RESUMEN

Infiltrating T cells and monocyte-derived macrophages support central nervous system repair. Although infiltration of leucocytes to the injured central nervous system has recently been shown to be orchestrated by the brain's choroid plexus, the immunological mechanism that maintains this barrier and regulates its activity as a selective gate is poorly understood. Here, we hypothesized that CD4(+) effector memory T cells, recently shown to reside at the choroid plexus stroma, regulate leucocyte trafficking through this portal through their interactions with the choroid plexus epithelium. We found that the naïve choroid plexus is populated by T helper 1, T helper 2 and regulatory T cells, but not by encephalitogenic T cells. In vitro findings revealed that the expression of immune cell trafficking determinants by the choroid plexus epithelium is specifically induced by interferon-γ. Tumour necrosis factor-α and interferon-γ reciprocally controlled the expression of their receptors by the choroid plexus epithelium, and had a synergistic effect in inducing the epithelial expression of trafficking molecules. In vivo, interferon-γ-dependent signalling controlled trafficking through the choroid plexus; interferon-γ receptor knockout mice exhibited reduced levels of T cells and monocyte entry to the cerebrospinal fluid and impaired recovery following spinal cord injury. Moreover, reduced expression of trafficking molecules by the choroid plexus was correlated with reduced CD4(+) T cells in the choroid plexus and cerebrospinal fluid of interferon-γ receptor knockout mice. Similar effect on the expression of trafficking molecules by the choroid plexus was found in bone-marrow chimeric mice lacking interferon-γ receptor in the central nervous system, or reciprocally, lacking interferon-γ in the circulation. Collectively, our findings attribute a novel immunological plasticity to the choroid plexus epithelium, allowing it to serve, through interferon-γ signalling, as a tightly regulated entry gate into the central nervous system for circulating leucocytes immune surveillance under physiological conditions, and for repair following acute injury.


Asunto(s)
Sistema Nervioso Central/inmunología , Plexo Coroideo/inmunología , Plexo Coroideo/patología , Interferón gamma/fisiología , Animales , Movimiento Celular/genética , Movimiento Celular/inmunología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Plexo Coroideo/metabolismo , Epitelio/inmunología , Epitelio/metabolismo , Epitelio/patología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transporte de Proteínas/genética , Transporte de Proteínas/inmunología , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Receptor de Interferón gamma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA