Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep Med ; 4(11): 101278, 2023 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-37944529

RESUMEN

The choroid plexus (CP) plays a key role in remotely controlling brain function in health, aging, and disease. Here, we report that CP epithelial cells express the brain-specific cholesterol 24-hydroxylase (CYP46A1) and that its levels are decreased under different mouse and human brain conditions, including amyloidosis, aging, and SARS-CoV-2 infection. Using primary mouse CP cell cultures, we demonstrate that the enzymatic product of CYP46A1, 24(S)-hydroxycholesterol, downregulates inflammatory transcriptomic signatures within the CP, found here to be elevated across multiple neurological conditions. In vitro, the pro-inflammatory cytokine tumor necrosis factor α (TNF-α) downregulates CYP46A1 expression, while overexpression of CYP46A1 or its pharmacological activation in mouse CP organ cultures increases resilience to TNF-α. In vivo, overexpression of CYP46A1 in the CP in transgenic mice with amyloidosis is associated with better cognitive performance and decreased brain inflammation. Our findings suggest that CYP46A1 expression in the CP impacts the role of this niche as a guardian of brain immune homeostasis.


Asunto(s)
Amiloidosis , Plexo Coroideo , Humanos , Ratones , Animales , Colesterol 24-Hidroxilasa/metabolismo , Plexo Coroideo/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Encéfalo/patología , Homeostasis/fisiología , Ratones Transgénicos , Amiloidosis/metabolismo , Amiloidosis/patología
2.
Nat Immunol ; 24(2): 220-224, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36717725

RESUMEN

The type I interferon (IFN) response is the body's typical immune defense against viruses. Previous studies linked high expression of genes encoding type I IFNs in the brain's choroid plexus to cognitive decline under virus-free conditions in aging and neurodegeneration. Multiple reports have documented persisting cognitive symptoms following recovery from COVID-19. Cumulative evidence shows that the choroid plexus is one of the brain regions most vulnerable to infection with the coronavirus SARS-CoV-2, and manifests increased expression of genes encoding type I IFNs even in the absence of viral traces within the brain. In this Perspective, we propose that the type I IFN defensive immune response to SARS-CoV-2 infection in the choroid plexus poses a risk to cognitive function if not resolved in a timely manner.


Asunto(s)
COVID-19 , Interferón Tipo I , Humanos , COVID-19/metabolismo , Interferón Tipo I/metabolismo , SARS-CoV-2/fisiología , Plexo Coroideo/metabolismo , Cognición , Antivirales/metabolismo , Interferones/metabolismo
3.
Neuron ; 110(21): 3421-3424, 2022 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-36150394

RESUMEN

Recent functional and anatomical discoveries of brain-immune relationships have overturned previous beliefs regarding the brain's immune privilege. Here, we propose that the brain and immune cells at its borders operate as an "ecosystem" to support the brain's robustness and resilience. Modulation of this ecosystem can be harnessed in the clinic.


Asunto(s)
Enfermedades Neurodegenerativas , Humanos , Enfermedades Neurodegenerativas/terapia , Encéfalo , Inmunoterapia
4.
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
5.
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
6.
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
7.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...