Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 42(3): 112196, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36871219

RESUMEN

The E4 allele of Apolipoprotein E (APOE) is associated with both metabolic dysfunction and a heightened pro-inflammatory response: two findings that may be intrinsically linked through the concept of immunometabolism. Here, we combined bulk, single-cell, and spatial transcriptomics with cell-specific and spatially resolved metabolic analyses in mice expressing human APOE to systematically address the role of APOE across age, neuroinflammation, and AD pathology. RNA sequencing (RNA-seq) highlighted immunometabolic changes across the APOE4 glial transcriptome, specifically in subsets of metabolically distinct microglia enriched in the E4 brain during aging or following an inflammatory challenge. E4 microglia display increased Hif1α expression and a disrupted tricarboxylic acid (TCA) cycle and are inherently pro-glycolytic, while spatial transcriptomics and mass spectrometry imaging highlight an E4-specific response to amyloid that is characterized by widespread alterations in lipid metabolism. Taken together, our findings emphasize a central role for APOE in regulating microglial immunometabolism and provide valuable, interactive resources for discovery and validation research.


Asunto(s)
Enfermedad de Alzheimer , Microglía , Ratones , Animales , Humanos , Microglía/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Apolipoproteína E4/metabolismo , Neuroglía/metabolismo , Encéfalo/metabolismo , Proteínas Amiloidogénicas/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Ratones Transgénicos , Apolipoproteína E3/metabolismo
2.
bioRxiv ; 2023 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-36798317

RESUMEN

Apolipoprotein E4 (APOE4) is the strongest risk allele associated with the development of late onset Alzheimer's disease (AD). Across the CNS, astrocytes are the predominant expressor of APOE while also being critical mediators of neuroinflammation and cerebral metabolism. APOE4 has been consistently linked with dysfunctional inflammation and metabolic processes, yet insights into the molecular constituents driving these responses remain unclear. Utilizing complementary approaches across humanized APOE mice and isogenic human iPSC astrocytes, we demonstrate that ApoE4 alters the astrocyte immunometabolic response to pro-inflammatory stimuli. Our findings show that ApoE4-expressing astrocytes acquire distinct transcriptional repertoires at single-cell and spatially-resolved domains, which are driven in-part by preferential utilization of the cRel transcription factor. Further, inhibiting cRel translocation in ApoE4 astrocytes abrogates inflammatory-induced glycolytic shifts and in tandem mitigates production of multiple pro-inflammatory cytokines. Altogether, our findings elucidate novel cellular underpinnings by which ApoE4 drives maladaptive immunometabolic responses of astrocytes.

3.
Mol Neurodegener ; 16(1): 62, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34488832

RESUMEN

BACKGROUND: Cerebral glucose hypometabolism is consistently observed in individuals with Alzheimer's disease (AD), as well as in young cognitively normal carriers of the Ε4 allele of Apolipoprotein E (APOE), the strongest genetic predictor of late-onset AD. While this clinical feature has been described for over two decades, the mechanism underlying these changes in cerebral glucose metabolism remains a critical knowledge gap in the field. METHODS: Here, we undertook a multi-omic approach by combining single-cell RNA sequencing (scRNAseq) and stable isotope resolved metabolomics (SIRM) to define a metabolic rewiring across astrocytes, brain tissue, mice, and human subjects expressing APOE4. RESULTS: Single-cell analysis of brain tissue from mice expressing human APOE revealed E4-associated decreases in genes related to oxidative phosphorylation, particularly in astrocytes. This shift was confirmed on a metabolic level with isotopic tracing of 13C-glucose in E4 mice and astrocytes, which showed decreased pyruvate entry into the TCA cycle and increased lactate synthesis. Metabolic phenotyping of E4 astrocytes showed elevated glycolytic activity, decreased oxygen consumption, blunted oxidative flexibility, and a lower rate of glucose oxidation in the presence of lactate. Together, these cellular findings suggest an E4-associated increase in aerobic glycolysis (i.e. the Warburg effect). To test whether this phenomenon translated to APOE4 humans, we analyzed the plasma metabolome of young and middle-aged human participants with and without the Ε4 allele, and used indirect calorimetry to measure whole body oxygen consumption and energy expenditure. In line with data from E4-expressing female mice, a subgroup analysis revealed that young female E4 carriers showed a striking decrease in energy expenditure compared to non-carriers. This decrease in energy expenditure was primarily driven by a lower rate of oxygen consumption, and was exaggerated following a dietary glucose challenge. Further, the stunted oxygen consumption was accompanied by markedly increased lactate in the plasma of E4 carriers, and a pathway analysis of the plasma metabolome suggested an increase in aerobic glycolysis. CONCLUSIONS: Together, these results suggest astrocyte, brain and system-level metabolic reprogramming in the presence of APOE4, a 'Warburg like' endophenotype that is observable in young females decades prior to clinically manifest AD.


Asunto(s)
Aerobiosis , Apolipoproteína E4/fisiología , Glucosa/metabolismo , Glucólisis , Síntomas Prodrómicos , Adolescente , Adulto , Anciano , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Apolipoproteína E4/genética , Astrocitos/metabolismo , Secuencia de Bases , Química Encefálica , Células Cultivadas , Diagnóstico Precoz , Metabolismo Energético , Femenino , Cromatografía de Gases y Espectrometría de Masas , Técnicas de Sustitución del Gen , Humanos , Metabolómica , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Oxidación-Reducción , Fosforilación Oxidativa , Consumo de Oxígeno/genética , Caracteres Sexuales , Análisis de la Célula Individual , Adulto Joven
4.
Exp Neurol ; 329: 113310, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32289316

RESUMEN

Innate immune responses, particularly activation of macrophages and microglia, are increasingly implicated in CNS disorders. It is now appreciated that the heterogeneity of functions adopted by these cells dictates neuropathophysiology. Research efforts to characterize the range of pro-inflammatory and anti-inflammatory phenotypes and functions adopted by microglia and macrophages are fueled by the potential for inflammatory cells to both exacerbate neurodegeneration and promote repair/disease resolution. The stimulation-based, M1/M2 classification system has emerged over the last decade as a common language to discuss macrophage and microglia heterogeneity across different fields. However, discontinuities between phenotypic markers and function create potential hurdles for the utility of the M1/M2 system in the development of effective immunomodulatory therapeutics for neuroinflammation. A framework to approach macrophage and microglia heterogeneity from a function-based phenotypic approach comes from rapidly emerging evidence that metabolic processes regulate immune cell activation. This concept of immunometabolism, however, is only beginning to unfold in the study of neurodegeneration and has yet to receive much focus in the context of neurotrauma. In this review, we first discuss the current views of macrophage and microglia heterogeneity and limitations of the M1/M2 classification system for neuropathological studies. We then review and discuss the current literature supporting metabolism as a regulator of microglia function in vitro. Lastly, we evaluate the evidence that metabolism regulates microglia and macrophage phenotype in vivo in models of Alzheimer's disease (AD), stroke, traumatic brain injury (TBI) and spinal cord injury (SCI).


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Inmunidad Celular/fisiología , Enfermedades Neurodegenerativas/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Animales , Lesiones Traumáticas del Encéfalo/inmunología , Enfermedades del Sistema Nervioso Central/inmunología , Enfermedades del Sistema Nervioso Central/metabolismo , Humanos , Enfermedades Neurodegenerativas/inmunología , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Traumatismos de la Médula Espinal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...