Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
3.
Brain Pathol ; 32(3): e13032, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34713522

RESUMEN

Several degenerative brain disorders such as Alzheimer's disease (AD), Parkinson's disease (PD) and Dementia with Lewy bodies (DLB) are characterized by the simultaneous appearance of amyloid-ß (Aß) and α-synuclein (α-syn) pathologies and symptoms that are similar, making it difficult to differentiate between these diseases. Until now, an accurate diagnosis can only be made by postmortem analysis. Furthermore, the role of α-syn in Aß aggregation and the arising characteristic olfactory impairments observed during the progression of these diseases is still not well understood. Therefore, we assessed Aß load in olfactory bulbs of APP-transgenic mice expressing APP695KM670/671NL and PSEN1L166P under the control of the neuron-specific Thy-1 promoter (referred to here as APPPS1) and APPPS1 mice co-expressing SNCAA30P (referred to here as APPPS1 × [A30P]aSYN). Furthermore, the olfactory capacity of these mice was evaluated in the buried food and olfactory avoidance test. Our results demonstrate an age-dependent increase in Aß load in the olfactory bulb of APP-transgenic mice that go along with exacerbated olfactory performance. Our study provides clear evidence that the presence of α-syn significantly diminished the endogenous and seed-induced Aß deposits and significantly ameliorated olfactory dysfunction in APPPS1 × [A30P]aSYN mice.


Asunto(s)
Enfermedad de Alzheimer , Sinucleinopatías , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Bulbo Olfatorio/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
4.
Nat Immunol ; 22(11): 1382-1390, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34663978

RESUMEN

Intergenerational inheritance of immune traits linked to epigenetic modifications has been demonstrated in plants and invertebrates. Here we provide evidence for transmission of trained immunity across generations to murine progeny that survived a sublethal systemic infection with Candida albicans or a zymosan challenge. The progeny of trained mice exhibited cellular, developmental, transcriptional and epigenetic changes associated with the bone marrow-resident myeloid effector and progenitor cell compartment. Moreover, the progeny of trained mice showed enhanced responsiveness to endotoxin challenge, alongside improved protection against systemic heterologous Escherichia coli and Listeria monocytogenes infections. Sperm DNA of parental male mice intravenously infected with the fungus C. albicans showed DNA methylation differences linked to immune gene loci. These results provide evidence for inheritance of trained immunity in mammals, enhancing protection against infections.


Asunto(s)
Candida albicans/inmunología , Candidiasis/inmunología , Infecciones por Escherichia coli/inmunología , Escherichia coli/inmunología , Herencia , Inmunidad Innata/genética , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Células Mieloides/inmunología , Animales , Candida albicans/patogenicidad , Candidiasis/genética , Candidiasis/metabolismo , Candidiasis/microbiología , Células Cultivadas , Metilación de ADN , Modelos Animales de Enfermedad , Epigénesis Genética , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Interacciones Huésped-Patógeno , Listeria monocytogenes/patogenicidad , Listeriosis/genética , Listeriosis/metabolismo , Listeriosis/microbiología , Masculino , Ratones Transgénicos , Células Mieloides/metabolismo , Células Mieloides/microbiología , Espermatozoides/inmunología , Espermatozoides/metabolismo , Transcripción Genética
5.
Brain Pathol ; 30(1): 36-45, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31099449

RESUMEN

Aggregation of amyloid-ß (Aß) that leads to the formation of plaques in Alzheimer's disease (AD) occurs through the stepwise formation of oligomers and fibrils. An earlier onset of aggregation is obtained upon intracerebral injection of Aß-containing brain homogenate into human APP transgenic mice that follows a prion-like seeding mechanism. Immunoprecipitation of these brain extracts with anti-Aß oligomer antibodies or passive immunization of the recipient animals abrogated the observed seeding activity, although induced Aß deposition was still evident. Here, we establish that, together with Aß monomers, Aß oligomers trigger the initial phase of Aß seeding and that the depletion of oligomeric Aß delays the aggregation process, leading to a transient reduction of seed-induced Aß deposits. This work extends the current knowledge about the role of Aß oligomers beyond its cytotoxic nature by pointing to a role in the initiation of Aß aggregation in vivo. We conclude that Aß oligomers are important for the early initiation phase of the seeding process.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Placa Amiloide/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Amiloide/metabolismo , Péptidos beta-Amiloides/fisiología , Animales , Encéfalo/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Placa Amiloide/fisiopatología , Agregación Patológica de Proteínas/metabolismo
6.
Methods Mol Biol ; 1989: 281-294, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31077112

RESUMEN

Recent advances in single cell multi-omics methodologies significantly expand our understanding of cellular heterogeneity, particularly in the field of immunology. Today's state-of-the-art flow and mass cytometers can detect up to 50 parameters to comprehensively characterize the identity and function of individual cells within a heterogeneous population. As a consequence, the increasing number of parameters that can be detected simultaneously also introduces substantial complexity for the experimental setup and downstream data processing. However, this challenge in data analysis fostered the development of novel bioinformatic tools to fully exploit the high-dimensional data. These tools will eventually replace cumbersome serial, manual gating in the two-dimensional space driven by a priori knowledge, which still represents the gold standard in flow cytometric analysis, to meet the needs of the today's immunologist. To this end, we provide guidelines for a high-dimensional cytometry workflow including experimental setup, panel design, fluorescent spillover compensation, and data analysis.


Asunto(s)
Algoritmos , Biología Computacional/métodos , Minería de Datos/métodos , Citometría de Flujo/métodos , Espectrometría de Masas/métodos , Análisis por Conglomerados , Humanos
7.
Nat Neurosci ; 22(2): 191-204, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30617257

RESUMEN

Coding variants in the triggering receptor expressed on myeloid cells 2 (TREM2) are associated with late-onset Alzheimer's disease (AD). We demonstrate that amyloid plaque seeding is increased in the absence of functional Trem2. Increased seeding is accompanied by decreased microglial clustering around newly seeded plaques and reduced plaque-associated apolipoprotein E (ApoE). Reduced ApoE deposition in plaques is also observed in brains of AD patients carrying TREM2 coding variants. Proteomic analyses and microglia depletion experiments revealed microglia as one origin of plaque-associated ApoE. Longitudinal amyloid small animal positron emission tomography demonstrates accelerated amyloidogenesis in Trem2 loss-of-function mutants at early stages, which progressed at a lower rate with aging. These findings suggest that in the absence of functional Trem2, early amyloidogenesis is accelerated due to reduced phagocytic clearance of amyloid seeds despite reduced plaque-associated ApoE.


Asunto(s)
Enfermedad de Alzheimer/genética , Amiloide/metabolismo , Apolipoproteínas E/metabolismo , Encéfalo/patología , Glicoproteínas de Membrana/genética , Placa Amiloide/genética , Receptores Inmunológicos/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Genotipo , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Transgénicos , Microglía/metabolismo , Microglía/patología , Fagocitosis/fisiología , Placa Amiloide/metabolismo , Placa Amiloide/patología , Receptores Inmunológicos/metabolismo
8.
EMBO J ; 37(2): 167-182, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29229786

RESUMEN

Alzheimer's disease (AD) is characterized by severe neuronal loss as well as the accumulation of amyloid-ß (Aß), which ultimately leads to plaque formation. Although there is now a general agreement that the aggregation of Aß can be initiated by prion-like seeding, the impact and functional consequences of induced Aß deposits (Aß seeding) on neurons still remain open questions. Here, we find that Aß seeding, representing early stages of plaque formation, leads to a dramatic decrease in proliferation and neurogenesis in two APP transgenic mouse models. We further demonstrate that neuronal cell death occurs primarily in the vicinity of induced Aß deposits culminating in electrophysiological abnormalities. Notably, environmental enrichment and voluntary exercise not only revives adult neurogenesis and reverses memory deficits but, most importantly, prevents Aß seeding by activated, phagocytic microglia cells. Our work expands the current knowledge regarding Aß seeding and the consequences thereof and attributes microglia an important role in diminishing Aß seeding by environmental enrichment.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Proliferación Celular , Microglía/metabolismo , Fagocitosis , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/genética , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Microglía/patología
9.
Immunity ; 47(6): 1051-1066.e12, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29262348

RESUMEN

Human in vitro generated monocyte-derived dendritic cells (moDCs) and macrophages are used clinically, e.g., to induce immunity against cancer. However, their physiological counterparts, ontogeny, transcriptional regulation, and heterogeneity remains largely unknown, hampering their clinical use. High-dimensional techniques were used to elucidate transcriptional, phenotypic, and functional differences between human in vivo and in vitro generated mononuclear phagocytes to facilitate their full potential in the clinic. We demonstrate that monocytes differentiated by macrophage colony-stimulating factor (M-CSF) or granulocyte macrophage colony-stimulating factor (GM-CSF) resembled in vivo inflammatory macrophages, while moDCs resembled in vivo inflammatory DCs. Moreover, differentiated monocytes presented with profound transcriptomic, phenotypic, and functional differences. Monocytes integrated GM-CSF and IL-4 stimulation combinatorically and temporally, resulting in a mode- and time-dependent differentiation relying on NCOR2. Finally, moDCs are phenotypically heterogeneous and therefore necessitate the use of high-dimensional phenotyping to open new possibilities for better clinical tailoring of these cellular therapies.


Asunto(s)
Células Dendríticas/inmunología , Interleucina-4/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Co-Represor 2 de Receptor Nuclear/inmunología , Transducción de Señal/inmunología , Diferenciación Celular , Linaje de la Célula , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Inmunofenotipificación , Interleucina-4/genética , Interleucina-4/farmacología , Activación de Macrófagos , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Monocitos/citología , Monocitos/efectos de los fármacos , Co-Represor 2 de Receptor Nuclear/genética , Cultivo Primario de Células , Factores de Tiempo , Transcripción Genética
10.
Sci Rep ; 5: 14624, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26416689

RESUMEN

The role of microglia in amyloid-ß (Aß) deposition is controversial. In the present study, an organotypic hippocampal slice culture (OHSC) system with an in vivo-like microglial-neuronal environment was used to investigate the potential contribution of microglia to Aß plaque formation. We found that microglia ingested Aß, thereby preventing plaque formation in OHSCs. Conversely, Aß deposits formed rapidly in microglia-free wild-type slices. The capacity to prevent Aß plaque formation was absent in forebrain microglia from young adult but not juvenile 5xFamilial Alzheimer's disease (FAD) mice. Since no loss of Aß clearance capacity was observed in both wild-type and cerebellar microglia from 5xFAD animals, the high Aß1-42 burden in the forebrain of 5xFAD animals likely underlies the exhaustion of microglial Aß clearance capacity. These data may therefore explain why Aß plaque formation has never been described in wild-type mice, and point to a beneficial role of microglia in AD pathology. We also describe a new method to study Aß plaque formation in a cell culture setting.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Hipocampo/patología , Microglía/patología , Fragmentos de Péptidos/metabolismo , Placa Amiloide/patología , Prosencéfalo/patología , Factores de Edad , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/prevención & control , Animales , Transporte Biológico , Modelos Animales de Enfermedad , Resistencia a la Enfermedad/fisiología , Femenino , Hipocampo/metabolismo , Hipocampo/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Microglía/ultraestructura , Microtomía , Placa Amiloide/metabolismo , Placa Amiloide/prevención & control , Prosencéfalo/metabolismo , Prosencéfalo/ultraestructura , Técnicas de Cultivo de Tejidos
11.
Nat Med ; 21(7): 802-7, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26099047

RESUMEN

Amyloid-ß (Aß) plaques and α-synuclein (α-syn)-rich Lewy bodies are the major neuropathological hallmarks of Alzheimer's disease (AD) and Parkinson's disease, respectively. An overlap of pathologies is found in most individuals with dementia with Lewy bodies (DLB) and in more than 50% of AD cases. Their brains display substantial α-syn accumulation not only in Lewy bodies, but also in dystrophic neurites decorating Aß plaques. Several studies report binding and coaggregation of Aß and α-syn, yet the precise role of α-syn in amyloid plaque formation remains elusive. Here we performed intracerebral injections of α-syn-containing preparations into amyloid precursor protein (APP) transgenic mice (expressing APP695(KM670/671NL) and PSEN1(L166P) under the control of the neuron-specific Thy-1 promoter; referred to here as 'APPPS1'). Unexpectedly, α-syn failed to cross-seed Aß plaques in vivo, but rather it inhibited plaque formation in APPPS1 mice coexpressing SNCA(A30P) (referred to here as 'APPPS1 × [A30P]aSYN' double-transgenic mice). This was accompanied by increased Aß levels in cerebrospinal fluid despite unchanged overall Aß levels. Notably, the seeding activity of Aß-containing brain homogenates was considerably reduced by α-syn, and Aß deposition was suppressed in grafted tissue from [A30P]aSYN transgenic mice. Thus, we conclude that an interaction between Aß and α-syn leads to inhibition of Aß deposition and to reduced plaque formation.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Placa Amiloide/metabolismo , alfa-Sinucleína/metabolismo , Péptidos beta-Amiloides/ultraestructura , Animales , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/patología , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Placa Amiloide/ultraestructura , Presenilina-1/metabolismo
12.
Acta Neuropathol ; 128(3): 333-45, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25056803

RESUMEN

Microglia, the tissue-resident macrophages of the brain, are attracting increasing attention as key players in brain homeostasis from development through aging. Recent works have highlighted new and unexpected roles for these once-enigmatic cells in both healthy central nervous system function and in diverse pathologies long thought to be primarily the result of neuronal malfunction. In this review, we have chosen to focus on Rett syndrome, which features early neurodevelopmental pathology, and Alzheimer's disease, a disorder associated predominantly with aging. Interestingly, receptor-mediated microglial phagocytosis has emerged as a key function in both developmental and late-life brain pathologies. In a mouse model of Rett syndrome, bone marrow transplant and CNS engraftment of microglia-like cells were associated with surprising improvements in pathology-these benefits were abrogated by block of phagocytic function. In Alzheimer's disease, large-scale genome-wide association studies have been brought to bear as a method of identifying previously unknown susceptibility genes, which highlight microglial receptors as promising novel targets for therapeutic modulation. Multi-photon in vivo microscopy has provided a method of directly visualizing the effects of manipulation of these target genes. Here, we review the latest findings and concepts emerging from the rapidly growing body of literature exemplified for Rett syndrome and late-onset, sporadic Alzheimer's disease.


Asunto(s)
Encéfalo , Enfermedades del Sistema Nervioso Central/patología , Microglía/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Encéfalo/citología , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Modelos Animales de Enfermedad , Humanos , Ratones
13.
PLoS One ; 8(3): e58544, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23526995

RESUMEN

The engraftment of bone marrow-derived cells has been described not only during diseases of the central nervous system (CNS) but also under healthy conditions. However, previous studies pointing to an ample bone marrow cell engraftment used irradiation-induced bone marrow chimeras that evoked severe alterations of the CNS micromilieu including disturbances of the blood brain barrier (BBB), damage of endothelial cells and local induction of proinflammatory cytokines. On the other hand, parabiosis experiments using temporarily joined circulatory systems generally yielded low levels of myeloid cell chimerism thereby potentially underestimating bone marrow cell turnover with the CNS. To avoid these drawbacks we established a protocol using the alkylating agent busulfan prior to allogenic bone marrow transplantation from CX3CR1(GFP/+) donors. This regimen resulted in a stable and high peripheral myeloid chimerism, significantly reduced cytokine induction and preserved BBB integrity. Importantly, bone marrow cell recruitment to the CNS was strongly diminished under these conditions and only weakly enhanced during local neurodegeneration induced by facial nerve axotomy. These results underscore the requirement of local CNS conditioning for efficient recruitment of bone marrow cells, establish busulfan as an alternative treatment for studying bone marrow chimeras and suggest a critical re-evaluation of earlier chimeric studies involving irradiation or parabiosis regimens.


Asunto(s)
Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Encéfalo/citología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Busulfano/farmacología , Movimiento Celular , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Agonistas Mieloablativos/farmacología , Parabiosis , Quimera por Radiación , Quimera por Trasplante , Acondicionamiento Pretrasplante , Trasplante Autólogo , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...