Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Mol Psychiatry ; 25(5): 1050-1067, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31772304

RESUMO

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.


Assuntos
Comportamento Animal , Interferon Tipo I/imunologia , Microglia/imunologia , Mães/psicologia , Efeitos Tardios da Exposição Pré-Natal/imunologia , Estresse Psicológico/imunologia , Animais , Modelos Animais de Doenças , Feminino , Exposição Materna , Camundongos , Camundongos Endogâmicos C57BL , Gravidez
2.
EMBO J ; 34(13): 1816-28, 2015 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-25940071

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Quimiotaxia de Leucócito/efeitos dos fármacos , Plexo Corióideo/efeitos dos fármacos , Leucócitos/efeitos dos fármacos , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico/farmacologia , Doença de Alzheimer/genética , Doença de Alzheimer/imunologia , Doença de Alzheimer/patologia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/imunologia , Células Cultivadas , Plexo Corióideo/imunologia , Plexo Corióideo/metabolismo , Modelos Animais de Doenças , Feminino , Leucócitos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
3.
Brain ; 136(Pt 11): 3427-40, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24088808

RESUMO

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.


Assuntos
Sistema Nervoso Central/imunologia , Plexo Corióideo/imunologia , Plexo Corióideo/patologia , Interferon gama/fisiologia , Animais , Movimento Celular/genética , Movimento Celular/imunologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Plexo Corióideo/metabolismo , Epitélio/imunologia , Epitélio/metabolismo , Epitélio/patologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transporte Proteico/genética , Transporte Proteico/imunologia , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Receptor de Interferon gama
4.
Cell Rep Med ; 5(5): 101546, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38703766

RESUMO

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.


Assuntos
Esclerose Lateral Amiotrófica , Fatores Inibidores da Migração de Macrófagos , Neurônios Motores , Superóxido Dismutase-1 , Fatores Inibidores da Migração de Macrófagos/metabolismo , Fatores Inibidores da Migração de Macrófagos/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/terapia , Esclerose Lateral Amiotrófica/patologia , Animais , Humanos , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo , Camundongos , Células-Tronco Pluripotentes Induzidas/metabolismo , Oxirredutases Intramoleculares/metabolismo , Oxirredutases Intramoleculares/genética , Camundongos Transgênicos , Dependovirus/genética , Modelos Animais de Doenças , Masculino , Mutação/genética , Feminino , Dobramento de Proteína
5.
Nat Commun ; 8(1): 717, 2017 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-28959042

RESUMO

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.


Assuntos
Envelhecimento/imunologia , Encéfalo/imunologia , Interferon beta/imunologia , Fatores de Transcrição MEF2/imunologia , Microglia/imunologia , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiopatologia , Humanos , Interferon beta/genética , Fatores de Transcrição MEF2/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL
6.
Nat Med ; 22(2): 135-7, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26779813

RESUMO

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.


Assuntos
Doença de Alzheimer/imunologia , Anticorpos/farmacologia , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Memória/efeitos dos fármacos , Placa Amiloide/imunologia , Receptor de Morte Celular Programada 1/imunologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/efeitos dos fármacos , Peptídeos beta-Amiloides/imunologia , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/imunologia , Encéfalo/patologia , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Interferon gama/efeitos dos fármacos , Interferon gama/imunologia , Macrófagos/imunologia , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Placa Amiloide/patologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Análise de Sequência de RNA , Transdução de Sinais/efeitos dos fármacos
7.
Nat Commun ; 6: 7967, 2015 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-26284939

RESUMO

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.


Assuntos
Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Linfócitos T Reguladores/metabolismo , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Giro Denteado/patologia , Giro Denteado/fisiologia , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica/fisiologia , Acetato de Glatiramer/farmacologia , Tolerância Imunológica , Imunomodulação , Camundongos , Camundongos Transgênicos , RNA/genética , RNA/metabolismo
8.
Science ; 346(6205): 89-93, 2014 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-25147279

RESUMO

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.


Assuntos
Envelhecimento/patologia , Encéfalo/fisiologia , Plexo Corióideo/metabolismo , Cognição , Regulação da Expressão Gênica , Fatores Reguladores de Interferon/genética , Interferon Tipo I/fisiologia , Envelhecimento/genética , Animais , Hipocampo/citologia , Camundongos , Camundongos Transgênicos , Neurogênese , Receptores de Interferon/genética , Receptor de Interferon gama
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA