Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Neurodegener ; 19(1): 42, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802940

RESUMEN

Microglia play diverse pathophysiological roles in Alzheimer's disease (AD), with genetic susceptibility factors skewing microglial cell function to influence AD risk. CD33 is an immunomodulatory receptor associated with AD susceptibility through a single nucleotide polymorphism that modulates mRNA splicing, skewing protein expression from a long protein isoform (CD33M) to a short isoform (CD33m). Understanding how human CD33 isoforms differentially impact microglial cell function in vivo has been challenging due to functional divergence of CD33 between mice and humans. We address this challenge by studying transgenic mice expressing either of the human CD33 isoforms crossed with the 5XFAD mouse model of amyloidosis and find that human CD33 isoforms have opposing effects on the response of microglia to amyloid-ß (Aß) deposition. Mice expressing CD33M have increased Aß levels, more diffuse plaques, fewer disease-associated microglia, and more dystrophic neurites compared to 5XFAD control mice. Conversely, CD33m promotes plaque compaction and microglia-plaque contacts, and minimizes neuritic plaque pathology, highlighting an AD protective role for this isoform. Protective phenotypes driven by CD33m are detected at an earlier timepoint compared to the more aggressive pathology in CD33M mice that appears at a later timepoint, suggesting that CD33m has a more prominent impact on microglia cell function at earlier stages of disease progression. In addition to divergent roles in modulating phagocytosis, scRNAseq and proteomics analyses demonstrate that CD33m+ microglia upregulate nestin, an intermediate filament involved in cell migration, at plaque contact sites. Overall, our work provides new functional insights into how CD33, as a top genetic susceptibility factor for AD, modulates microglial cell function.


Asunto(s)
Enfermedad de Alzheimer , Modelos Animales de Enfermedad , Ratones Transgénicos , Microglía , Isoformas de Proteínas , Lectina 3 Similar a Ig de Unión al Ácido Siálico , Animales , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Microglía/metabolismo , Lectina 3 Similar a Ig de Unión al Ácido Siálico/metabolismo , Humanos , Ratones , Isoformas de Proteínas/metabolismo , Péptidos beta-Amiloides/metabolismo , Placa Amiloide/metabolismo , Placa Amiloide/patología
2.
Mol Neurodegener ; 16(1): 19, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33766097

RESUMEN

BACKGROUND: CD33 is genetically linked to Alzheimer's disease (AD) susceptibility through differential expression of isoforms in microglia. The role of the human CD33 short isoform (hCD33m), preferentially encoded by an AD-protective CD33 allele (rs12459419T), is unknown. Here, we test whether hCD33m represents a loss-of-function or gain-of-function variant. METHODS: We have developed two models to test the role of hCD33m. The first is a new strain of transgenic mice expressing hCD33m in the microglial cell lineage. The second is U937 cells where the CD33 gene was disrupted by CRISPR/Cas9 and complemented with different variants of hCD33. Primary microglia and U937 cells were tested in phagocytosis assays and single cell RNA sequencing (scRNAseq) was carried out on the primary microglia. Furthermore, a new monoclonal antibody was developed to detect hCD33m more efficiently. RESULTS: In both primary microglia and U937 cells, we find that hCD33m enhances phagocytosis. This contrasts with the human CD33 long isoform (hCD33M) that represses phagocytosis, as previously demonstrated. As revealed by scRNAseq, hCD33m+ microglia are enriched in a cluster of cells defined by an upregulated expression and gene regulatory network of immediate early genes, which was further validated within microglia in situ. Using a new hCD33m-specific antibody enabled hCD33m expression to be examined, demonstrating a preference for an intracellular location. Moreover, this newly discovered gain-of-function role for hCD33m is dependent on its cytoplasmic signaling motifs, dominant over hCD33M, and not due to loss of glycan ligand binding. CONCLUSIONS: These results provide strong support that hCD33m represents a gain-of-function isoform and offers insight into what it may take to therapeutically capture the AD-protective CD33 allele.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Microglía/fisiología , Fragmentos de Péptidos/metabolismo , Fagocitosis/genética , Lectina 3 Similar a Ig de Unión al Ácido Siálico/genética , Alelos , Animales , Sistemas CRISPR-Cas , Cruzamientos Genéticos , Femenino , Mutación con Ganancia de Función , Edición Génica , Redes Reguladoras de Genes , Genes Inmediatos-Precoces , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Polisacáridos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , RNA-Seq , Lectina 3 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Lectina 3 Similar a Ig de Unión al Ácido Siálico/fisiología , Análisis de la Célula Individual , Células U937
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...