Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Nat Immunol ; 24(11): 1839-1853, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37749326

RESUMO

The APOE4 allele is the strongest genetic risk factor for late-onset Alzheimer's disease (AD). The contribution of microglial APOE4 to AD pathogenesis is unknown, although APOE has the most enriched gene expression in neurodegenerative microglia (MGnD). Here, we show in mice and humans a negative role of microglial APOE4 in the induction of the MGnD response to neurodegeneration. Deletion of microglial APOE4 restores the MGnD phenotype associated with neuroprotection in P301S tau transgenic mice and decreases pathology in APP/PS1 mice. MGnD-astrocyte cross-talk associated with ß-amyloid (Aß) plaque encapsulation and clearance are mediated via LGALS3 signaling following microglial APOE4 deletion. In the brains of AD donors carrying the APOE4 allele, we found a sex-dependent reciprocal induction of AD risk factors associated with suppression of MGnD genes in females, including LGALS3, compared to individuals homozygous for the APOE3 allele. Mechanistically, APOE4-mediated induction of ITGB8-transforming growth factor-ß (TGFß) signaling impairs the MGnD response via upregulation of microglial homeostatic checkpoints, including Inpp5d, in mice. Deletion of Inpp5d in microglia restores MGnD-astrocyte cross-talk and facilitates plaque clearance in APP/PS1 mice. We identify the microglial APOE4-ITGB8-TGFß pathway as a negative regulator of microglial response to AD pathology, and restoring the MGnD phenotype via blocking ITGB8-TGFß signaling provides a promising therapeutic intervention for AD.


Assuntos
Doença de Alzheimer , Feminino , Camundongos , Humanos , Animais , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Microglia/metabolismo , Galectina 3/genética , Galectina 3/metabolismo , Peptídeos beta-Amiloides/metabolismo , Camundongos Transgênicos , Modelos Animais de Doenças
2.
Nat Commun ; 10(1): 465, 2019 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-30692527

RESUMO

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.


Assuntos
Antígeno B7-H1/metabolismo , Disfunção Cognitiva/metabolismo , Macrófagos/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Tauopatias/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Anticorpos Bloqueadores/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Encéfalo/imunologia , Encéfalo/metabolismo , Disfunção Cognitiva/genética , Modelos Animais de Doenças , Humanos , Macrófagos/imunologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Transgênicos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Tauopatias/genética
3.
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
4.
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
5.
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
6.
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
7.
Immunity ; 38(3): 555-69, 2013 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-23477737

RESUMO

Monocyte-derived macrophages are essential for recovery after spinal cord injury, but their homing mechanism is poorly understood. Here, we show that although of common origin, the homing of proinflammatory (M1) and the "alternatively activated" anti-inflammatory (M2) macrophages to traumatized spinal cord (SC) was distinctly regulated, neither being through breached blood-brain barrier. The M1 macrophages (Ly6c(hi)CX3CR1(lo)) derived from monocytes homed in a CCL2 chemokine-dependent manner through the adjacent SC leptomeninges. The resolving M2 macrophages (Ly6c(lo)CX3CR1(hi)) derived from monocytes trafficked through a remote blood-cerebrospinal-fluid (CSF) barrier, the brain-ventricular choroid plexus (CP), via VCAM-1-VLA-4 adhesion molecules and epithelial CD73 enzyme for extravasation and epithelial transmigration. Blockage of these determinants, or mechanical CSF flow obstruction, inhibited M2 macrophage recruitment and impaired motor-function recovery. The CP, along with the CSF and the central canal, provided an anti-inflammatory supporting milieu, potentially priming the trafficking monocytes. Overall, our finding demonstrates that the route of monocyte entry to central nervous system provides an instructional environment to shape their function.


Assuntos
Plexo Corióideo/imunologia , Macrófagos/imunologia , Traumatismos da Medula Espinal/imunologia , Medula Espinal/imunologia , 5'-Nucleotidase/antagonistas & inibidores , 5'-Nucleotidase/genética , 5'-Nucleotidase/imunologia , Difosfato de Adenosina/análogos & derivados , Difosfato de Adenosina/farmacologia , Animais , Antígenos Ly/imunologia , Antígenos Ly/metabolismo , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Receptor 1 de Quimiocina CX3C , Movimento Celular/genética , Movimento Celular/imunologia , Plexo Corióideo/metabolismo , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Expressão Gênica/imunologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Integrina alfa4beta1/genética , Integrina alfa4beta1/imunologia , Antígenos Comuns de Leucócito/imunologia , Antígenos Comuns de Leucócito/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Meninges/imunologia , Meninges/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/metabolismo , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medula Espinal/metabolismo , Traumatismos da Medula Espinal/líquido cefalorraquidiano , Traumatismos da Medula Espinal/genética , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA