Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 169(7): 1276-1290.e17, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28602351

RESUMEN

Alzheimer's disease (AD) is a detrimental neurodegenerative disease with no effective treatments. Due to cellular heterogeneity, defining the roles of immune cell subsets in AD onset and progression has been challenging. Using transcriptional single-cell sorting, we comprehensively map all immune populations in wild-type and AD-transgenic (Tg-AD) mouse brains. We describe a novel microglia type associated with neurodegenerative diseases (DAM) and identify markers, spatial localization, and pathways associated with these cells. Immunohistochemical staining of mice and human brain slices shows DAM with intracellular/phagocytic Aß particles. Single-cell analysis of DAM in Tg-AD and triggering receptor expressed on myeloid cells 2 (Trem2)-/- Tg-AD reveals that the DAM program is activated in a two-step process. Activation is initiated in a Trem2-independent manner that involves downregulation of microglia checkpoints, followed by activation of a Trem2-dependent program. This unique microglia-type has the potential to restrict neurodegeneration, which may have important implications for future treatment of AD and other neurodegenerative diseases. VIDEO ABSTRACT.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Microglía/patología , Fagocitos/patología , Enfermedad de Alzheimer/genética , Animales , Humanos , Ratones , Ratones Transgénicos , Microglía/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/patología , Fagocitos/metabolismo , Receptores Inmunológicos/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual
2.
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
3.
Trends Immunol ; 37(3): 181-192, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26877243

RESUMEN

Recent findings have revealed distinct roles for type I and II interferons (IFN-I and IFN-γ) in the recruitment of immune cells to the central nervous system (CNS) and highlighted the importance of this process for brain maintenance and protection/repair. Furthermore, manipulation of IFN-I and IFN-γ pathways in pathological contexts has yielded conflicting results. We discuss these findings, focusing on two distinct conditions; relapsing remitting multiple sclerosis (RRMS) and brain aging. Using these examples, we propose that regulation of immune cell entry to the CNS is a mechanism through which interaction between IFN-I and -II can affect brain function from its anatomical borders. Deviation from homeostatic IFN-I/-II balance may contribute to distinct brain pathologies, resulting from either insufficient immune surveillance of the CNS and loss of immune-dependent protection, or overwhelming leukocyte entry and immune-mediated destruction.


Asunto(s)
Envejecimiento/inmunología , Encéfalo/fisiología , Interferón Tipo I/fisiología , Interferón gamma/fisiología , Esclerosis Múltiple Recurrente-Remitente/inmunología , Animales , Autoinmunidad , Homeostasis , Humanos , Vigilancia Inmunológica
4.
EMBO J ; 33(1): 7-22, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24357543

RESUMEN

Inflammation is an integral part of the body's physiological repair mechanism, unless it remains unresolved and becomes pathological, as evident in the progressive nature of neurodegeneration. Based on studies from outside the central nervous system (CNS), it is now understood that the resolution of inflammation is an active process, which is dependent on well-orchestrated innate and adaptive immune responses. Due to the immunologically privileged status of the CNS, such resolution mechanism has been mostly ignored. Here, we discuss resolution of neuroinflammation as a process that depends on a network of immune cells operating in a tightly regulated sequence, involving the brain's choroid plexus (CP), a unique neuro-immunological interface, positioned to integrate signals it receives from the CNS parenchyma with signals coming from circulating immune cells, and to function as an on-alert gate for selective recruitment of inflammation-resolving leukocytes to the inflamed CNS parenchyma. Finally, we propose that functional dysregulation of the CP reflects a common underlying mechanism in the pathophysiology of neurodegenerative diseases, and can thus serve as a potential novel target for therapy.


Asunto(s)
Encefalopatías/inmunología , Encefalopatías/patología , Plexo Coroideo/inmunología , Inflamación/inmunología , Inflamación/patología , Leucocitos/inmunología , Enfermedades Neurodegenerativas/fisiopatología , Animales , Humanos
5.
J Neurosci ; 35(16): 6381-93, 2015 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-25904790

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a devastating fatal motor neuron disease, for which there is currently no cure or effective treatment. In this disease, local neuroinflammation develops along the disease course and contributes to its rapid progression. In several models of CNS pathologies, circulating immune cells were shown to display an indispensable role in the resolution of the neuroinflammatory response. The recruitment of such cells to the CNS involves activation of the choroid plexus (CP) of the brain for leukocyte trafficking, through a mechanism that requires IFN-γ signaling. Here, we found that in the mutant SOD1(G93A) (mSOD1) mouse model of ALS, the CP does not support leukocyte trafficking during disease progression, due to a local reduction in IFN-γ levels. Therapeutic immunization of mSOD1 mice with a myelin-derived peptide led to CP activation, and was followed by the accumulation of immunoregulatory cells, including IL-10-producing monocyte-derived macrophages and Foxp3(+) regulatory T cells, and elevation of the neurotrophic factors IGF-1 and GDNF in the diseased spinal cord parenchyma. The immunization resulted in the attenuation of disease progression and an increased life expectancy of the mSOD1 mice. Collectively, our results demonstrate that recruitment of immunoregulatory cells to the diseased spinal cord in ALS, needed for fighting off the pathology, can be enhanced by transiently boosting peripheral immunity to myelin antigens.


Asunto(s)
Esclerosis Amiotrófica Lateral/inmunología , Plexo Coroideo/citología , Plexo Coroideo/inmunología , Progresión de la Enfermedad , Inmunización , Glicoproteína Mielina-Oligodendrócito/inmunología , Linfocitos T/inmunología , Esclerosis Amiotrófica Lateral/patología , Animales , Movimiento Celular/inmunología , Modelos Animales de Enfermedad , Femenino , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Macrófagos/citología , Macrófagos/inmunología , Masculino , Ratones , Ratones Transgénicos , Mutación , Cultivo Primario de Células , Médula Espinal/inmunología , Médula Espinal/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Linfocitos T/metabolismo
6.
Proc Natl Acad Sci U S A ; 110(6): 2264-9, 2013 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-23335631

RESUMEN

The adaptive arm of the immune system has been suggested as an important factor in brain function. However, given the fact that interactions of neurons or glial cells with T lymphocytes rarely occur within the healthy CNS parenchyma, the underlying mechanism is still a mystery. Here we found that at the interface between the brain and blood circulation, the epithelial layers of the choroid plexus (CP) are constitutively populated with CD4(+) effector memory cells with a T-cell receptor repertoire specific to CNS antigens. With age, whereas CNS specificity in this compartment was largely maintained, the cytokine balance shifted in favor of the T helper type 2 (Th2) response; the Th2-derived cytokine IL-4 was elevated in the CP of old mice, relative to IFN-γ, which decreased. We found this local cytokine shift to critically affect the CP epithelium, triggering it to produce the chemokine CCL11 shown to be associated with cognitive dysfunction. Partial restoration of cognitive ability in aged mice, by lymphopenia-induced homeostasis-driven proliferation of memory T cells, was correlated with restoration of the IL-4:IFN-γ ratio at the CP and modulated the expression of plasticity-related genes at the hippocampus. Our data indicate that the cytokine milieu at the CP epithelium is affected by peripheral immunosenescence, with detrimental consequences to the aged brain. Amenable to immunomodulation, this interface is a unique target for arresting age-related cognitive decline.


Asunto(s)
Envejecimiento/inmunología , Envejecimiento/patología , Encéfalo/inmunología , Encéfalo/patología , Plexo Coroideo/inmunología , Plexo Coroideo/patología , Células Th2/inmunología , Células Th2/patología , Inmunidad Adaptativa , Animales , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/patología , Proliferación Celular , Epitelio/inmunología , Epitelio/patología , Hipocampo/inmunología , Hipocampo/patología , Memoria Inmunológica , Linfopenia/inmunología , Linfopenia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroinmunomodulación , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Receptor de Interferón gamma
7.
J Autoimmun ; 60: 40-50, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25911200

RESUMEN

Neuropsychiatric disease is one of the most common manifestations of human systemic lupus erythematosus, but the mechanisms remain poorly understood. In human brain microvascular endothelial cells in vitro, TNF-like weak inducer of apoptosis (TWEAK) decreases tight junction ZO-1 expression and increases the permeability of monolayer cell cultures. Furthermore, knockout (KO) of the TWEAK receptor, Fn14, in the MRL/lpr lupus mouse strain markedly attenuates neuropsychiatric disease, as demonstrated by significant reductions in depressive-like behavior and improved cognitive function. The purpose of the present study was to determine the mechanisms by which TWEAK signaling is instrumental in the pathogenesis of neuropsychiatric lupus (NPSLE). Evaluating brain sections of MRL/lpr Fn14WT and Fn14KO mice, we found that Fn14KO mice displayed significantly decreased cellular infiltrates in the choroid plexus. To evaluate the integrity of the blood brain barrier (BBB) in MRL/lpr mice, Western blot for fibronectin, qPCR for iNOS, and immunohistochemical staining for VCAM-1/ICAM-1 were performed. We found preserved BBB permeability in MRL/lpr Fn14KO mice, attributable to reduced brain expression of VCAM-1/ICAM-1 and iNOS. Additionally, administration of Fc-TWEAK intravenously directly increased the leakage of a tracer (dextran-FITC) into brain tissue. Furthermore, MRL/lpr Fn14KO mice displayed reduced antibody (IgG) and complement (C3, C6, and C4a) deposition in the brain. Finally, we found that MRL/lpr Fn14KO mice manifested reduced neuron degeneration and hippocampal gliosis. Our studies indicate that TWEAK/Fn14 interactions play an important role in the pathogenesis of NPSLE by increasing the accumulation of inflammatory cells in the choroid plexus, disrupting BBB integrity, and increasing neuronal damage, suggesting a novel target for therapy in this disease.


Asunto(s)
Apoptosis/genética , Barrera Hematoencefálica/fisiopatología , Neuronas/patología , Receptores del Factor de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/inmunología , Animales , Apoptosis/inmunología , Plexo Coroideo/fisiopatología , Cognición , Complemento C3/inmunología , Complemento C4a/inmunología , Complemento C6/inmunología , Citocina TWEAK , Depresión/genética , Modelos Animales de Enfermedad , Gliosis/genética , Inmunoglobulina G/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Noqueados , Degeneración Nerviosa/genética , Permeabilidad , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor de TWEAK , Molécula 1 de Adhesión Celular Vascular/metabolismo , Proteína de la Zonula Occludens-1/biosíntesis
8.
J Autoimmun ; 54: 8-14, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25199710

RESUMEN

Immune cell infiltration to the brain's territory was considered for decades to reflect a pathological process in which immune cells attack the central nervous system (CNS); such a process is observed in the inflammatory autoimmune disease, multiple sclerosis (MS). As neuroinflammatory processes within the CNS parenchyma are also common to other CNS pathologies, regardless of their etiology, including neurodegenerative disorders such as Alzheimer's disease (AD) and Amyotrophic lateral sclerosis (ALS), these pathologies have often been compared to MS, a disease that benefits from immunosuppressive therapy. Yet, over the last decade, it became clear that autoimmunity has a bright side, and that it plays a pivotal role in CNS repair following damage. Specifically, autoimmune T cells were found to facilitate CNS healing processes, such as in the case of sterile mechanical injuries to the brain or the spinal cord, mental stress, or biochemical insults. Even more intriguingly, autoimmune T cells were found to be involved in supporting fundamental processes of brain functional integrity, such as in the maintenance of life-long brain plasticity, including spatial learning and memory, and neurogenesis. Importantly, autoimmune T cells are part of a cellular network which, to operate efficiently and safely, requires tight regulation by other immune cell populations, such as regulatory T cells, which are indispensable for maintenance of immunological self-tolerance and homeostasis. Here, we suggest that dysregulation of the balance between peripheral immune suppression, on one hand, and protective autoimmunity, on the other, is an underlying mechanism in the emergence and progression of the neuroinflammatory response associated with chronic neurodegenerative diseases and brain aging. Mitigating chronic neuroinflammation under these conditions necessitates activation, rather than suppression, of the peripheral immune response directed against self. Accordingly, we propose that fighting off acute and chronic neurodegenerative conditions requires breaking peripheral immune tolerance to CNS self-antigens, in order to boost protective autoimmunity. Nevertheless, the optimal approach to fine tune such immune response must be individually explored for each condition.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Esclerosis Amiotrófica Lateral/inmunología , Autoantígenos/inmunología , Tolerancia Inmunológica , Esclerosis Múltiple/inmunología , Linfocitos T Reguladores/inmunología , Enfermedad Aguda , Enfermedad de Alzheimer/patología , Esclerosis Amiotrófica Lateral/patología , Animales , Encéfalo/inmunología , Encéfalo/patología , Enfermedad Crónica , Humanos , Esclerosis Múltiple/patología , Médula Espinal/inmunología , Médula Espinal/patología , Linfocitos T Reguladores/patología
9.
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
10.
Front Immunol ; 15: 1343900, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38720902

RESUMEN

Alzheimer's disease has an increasing prevalence in the population world-wide, yet current diagnostic methods based on recommended biomarkers are only available in specialized clinics. Due to these circumstances, Alzheimer's disease is usually diagnosed late, which contrasts with the currently available treatment options that are only effective for patients at an early stage. Blood-based biomarkers could fill in the gap of easily accessible and low-cost methods for early diagnosis of the disease. In particular, immune-based blood-biomarkers might be a promising option, given the recently discovered cross-talk of immune cells of the central nervous system with those in the peripheral immune system. Here, we give a background on recent advances in research on brain-immune system cross-talk in Alzheimer's disease and review machine learning approaches, which can combine multiple biomarkers with further information (e.g. age, sex, APOE genotype) into predictive models supporting an earlier diagnosis. In addition, mechanistic modeling approaches, such as agent-based modeling open the possibility to model and analyze cell dynamics over time. This review aims to provide an overview of the current state of immune-system related blood-based biomarkers and their potential for the early diagnosis of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Biomarcadores , Diagnóstico Precoz , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/sangre , Humanos , Biomarcadores/sangre , Aprendizaje Automático , Animales
11.
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
12.
Brain Behav Immun ; 34: 11-6, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23597431

RESUMEN

Adaptive immunity was repeatedly shown to play a role in maintaining lifelong brain function. Under physiological conditions, this activity was associated with CD4+ T cells specific for brain self-antigens. Nevertheless, direct interactions of T cells with the healthy neuronal parenchyma are hardly detectable. Recent studies have identified the brain's choroid plexus (CP) as an active neuro-immunological interface, enriched with CNS-specific CD4+ T cells. Strategically positioned for receiving signals from both the central nervous system (CNS) through the cerebrospinal fluid (CSF), and from the circulation through epithelium-immune cell interactions, the CP has recently been recognized as an important immunological compartment in maintaining and restoring brain homeostasis/allostasis. Here, we propose that CNS-specific T cells shape brain function via the CP, and suggest this immunological control to be lost as part of aging, in general, and immune senescence, in particular. Accordingly, the CP may serve as a novel target for immunomodulation to restore brain equilibrium.


Asunto(s)
Encéfalo/inmunología , Linfocitos T CD4-Positivos/inmunología , Plexo Coroideo/inmunología , Envejecimiento/inmunología , Animales , Humanos , Ratones
13.
Nat Aging ; 2(1): 60-73, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-37118355

RESUMEN

Microglia and monocyte-derived macrophages (MDM) are key players in dealing with Alzheimer's disease. In amyloidosis mouse models, activation of microglia was found to be TREM2 dependent. Here, using Trem2-/-5xFAD mice, we assessed whether MDM act via a TREM2-dependent pathway. We adopted a treatment protocol targeting the programmed cell death ligand-1 (PD-L1) immune checkpoint, previously shown to modify Alzheimer's disease via MDM involvement. Blockade of PD-L1 in Trem2-/-5xFAD mice resulted in cognitive improvement and reduced levels of water-soluble amyloid beta1-42 with no effect on amyloid plaque burden. Single-cell RNA sequencing revealed that MDM, derived from both Trem2-/- and Trem2+/+5xFAD mouse brains, express a unique set of genes encoding scavenger receptors (for example, Mrc1, Msr1). Blockade of monocyte trafficking using anti-CCR2 antibody completely abrogated the cognitive improvement induced by anti-PD-L1 treatment in Trem2-/-5xFAD mice and similarly, but to a lesser extent, in Trem2+/+5xFAD mice. These results highlight a TREM2-independent, disease-modifying activity of MDM in an amyloidosis mouse model.


Asunto(s)
Enfermedad de Alzheimer , Amiloidosis , Ratones , Animales , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Ratones Transgénicos , Macrófagos/metabolismo , Amiloidosis/genética , Glicoproteínas de Membrana/genética , Receptores Inmunológicos/genética
14.
Int J Neuropsychopharmacol ; 12(7): 885-94, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19154657

RESUMEN

Multiple studies have reported oligodendrocyte and myelin abnormalities, as well as dysregulation of their related genes, in brains of schizophrenia patients. One of these genes is the myelin-basic-protein (MBP) gene, which encodes two families of proteins: classic-MBPs and golli-MBPs. While the classic-MBPs are predominantly located in the myelin sheaths of the nervous system, the golli proteins are more widely expressed and are found in both the immune and the nervous systems. In the present study we performed a case-control association analysis of golli-MBP in two separate Jewish Ashkenazi cohorts (cohort I: 120 patients, 236 controls; cohort II: 379 patients, 380 controls). In addition we performed an expression analysis of golli-MBP mRNA in post-mortem dorsolateral prefrontal cortex samples of schizophrenia patients, and matched controls. In the first cohort we observed association between six (out of 26 genotyped) single nucleotide polymorphisms (SNPs) and the disease (p<0.05). Of these, three are from one linkage disequilibrium (LD) block which contains a CTCF binding region. Haplotype analysis revealed significant 'risk'/'protective' haplotypes (strongest p=0.005, each) for schizophrenia. The three SNPs (rs12458282, rs2008323, rs721286) were then genotyped in the second cohort. The combined results showed strong effects, both in the single marker and in haplotype analyses (strongest OR 1.77, p=0.0005; OR 1.61, p=0.00001, respectively). Sequencing the CTCF binding region revealed three SNPs in complete LD with the associated haplotypes, located in close proximity to the CTCF binding site. Expression analysis found no significant differences in golli-MBP mRNA levels. These findings suggest that golli-MBP is a possible susceptibility gene for schizophrenia.


Asunto(s)
Judíos/genética , Judíos/psicología , Proteína Básica de Mielina/genética , Polimorfismo de Nucleótido Simple , Secuencias Reguladoras de Ácidos Nucleicos , Esquizofrenia/genética , Psicología del Esquizofrénico , Adulto , Anciano , Autopsia , Estudios de Casos y Controles , Distribución de Chi-Cuadrado , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Haplotipos , Humanos , Israel/epidemiología , Desequilibrio de Ligamiento , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Fenotipo , Corteza Prefrontal/química , ARN Mensajero/análisis , Medición de Riesgo , Factores de Riesgo , Esquizofrenia/etnología , Adulto Joven
15.
Anal Biochem ; 391(2): 91-7, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19464249

RESUMEN

Gene expression studies using postmortem human brain tissue are a common tool for studying the etiology of psychiatric disorders. Quantitative real-time PCR (qPCR) is an accurate and sensitive technique used for gene expression analysis in which the expression level is quantified by normalization to one or more reference genes. Therefore, accurate data normalization is critical for validating results obtained by qPCR. This study aimed to identify genes that may serve as reference in postmortem dorsolateral-prefrontal cortices (Brodmann's area 46) of schizophrenics, bipolar disorder (BPD) patients, and control subjects. In the exploratory stage of the analysis, samples of four BPD patients, two schizophrenics, and two controls were quantified using the TaqMan Low Density Array endogenous control panel, containing assays for 16 commonly used reference genes. In the next stage, six of these genes (TFRC, RPLP0, ACTB, POLR2a, B2M, and GAPDH) were quantified by qPCR in 12 samples of each clinical group. Expressional stability of the genes was determined by GeNorm and NormFinder. TFRC and RPLP0 were the most stably expressed genes, whereas the commonly used 18S, POLR2a, and GAPDH were the least stable. This report stresses the importance of examining expressional stability of candidate reference genes in the specific sample collection to be analyzed.


Asunto(s)
Trastorno Bipolar/genética , Expresión Génica , Reacción en Cadena de la Polimerasa/normas , Esquizofrenia/genética , Biomarcadores/análisis , Humanos , Reacción en Cadena de la Polimerasa/métodos , Estándares de Referencia
16.
Nat Commun ; 10(1): 465, 2019 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-30692527

RESUMEN

Alzheimer's disease (AD) is a heterogeneous disorder with multiple etiologies. Harnessing the immune system by blocking the programmed cell death receptor (PD)-1 pathway in an amyloid beta mouse model was shown to evoke a sequence of immune responses that lead to disease modification. Here, blocking PD-L1, a PD-1 ligand, was found to have similar efficacy to that of PD-1 blocking in disease modification, in both animal models of AD and of tauopathy. Targeting PD-L1 in a tau-driven disease model resulted in increased immunomodulatory monocyte-derived macrophages within the brain parenchyma. Single cell RNA-seq revealed that the homing macrophages expressed unique scavenger molecules including macrophage scavenger receptor 1 (MSR1), which was shown here to be required for the effect of PD-L1 blockade in disease modification. Overall, our results demonstrate that immune checkpoint blockade targeting the PD-1/PD-L1 pathway leads to modification of common factors that go awry in AD and dementia, and thus can potentially provide an immunotherapy to help combat these diseases.


Asunto(s)
Antígeno B7-H1/metabolismo , Disfunción Cognitiva/metabolismo , Macrófagos/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Tauopatías/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Anticuerpos Bloqueadores/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Encéfalo/inmunología , Encéfalo/metabolismo , Disfunción Cognitiva/genética , Modelos Animales de Enfermedad , Humanos , Macrófagos/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Transgénicos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Tauopatías/genética
17.
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
18.
Cell Stem Cell ; 18(6): 689-691, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27257756

RESUMEN

Hippocampal neurogenesis is affected throughout life by factors external to the brain, including what we eat, our gut microbiota, and the immune system. However, the mechanisms that link microbiota to neurogenesis are still puzzling. Now in Cell Reports, Möhle et al. (2016) attribute a role to Ly6C(hi) monocytes in this gut-immune-brain axis.


Asunto(s)
Tracto Gastrointestinal/inmunología , Monocitos/inmunología , Encéfalo , Humanos , Microbiota , Neurogénesis
19.
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
20.
Science ; 353(6301): aad8670, 2016 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-27338705

RESUMEN

Microglia, the resident myeloid cells of the central nervous system, play important roles in life-long brain maintenance and in pathology. Despite their importance, their regulatory dynamics during brain development have not been fully elucidated. Using genome-wide chromatin and expression profiling coupled with single-cell transcriptomic analysis throughout development, we found that microglia undergo three temporal stages of development in synchrony with the brain--early, pre-, and adult microglia--which are under distinct regulatory circuits. Knockout of the gene encoding the adult microglia transcription factor MAFB and environmental perturbations, such as those affecting the microbiome or prenatal immune activation, led to disruption of developmental genes and immune response pathways. Together, our work identifies a stepwise microglia developmental program integrating immune response pathways that may be associated with several neurodevelopmental disorders.


Asunto(s)
Encéfalo/embriología , Homeostasis/fisiología , Microglía/citología , Neurogénesis/inmunología , Animales , Barrera Hematoencefálica/embriología , Barrera Hematoencefálica/inmunología , Encéfalo/inmunología , Cromatina/metabolismo , Epigénesis Genética , Femenino , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Código de Histonas , Homeostasis/genética , Inmunidad/genética , Factor de Transcripción MafB/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/inmunología , Células Mieloides/citología , Neurogénesis/genética , Análisis de la Célula Individual
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA