Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Acta Neuropathol ; 142(5): 807-825, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34453582

RESUMEN

APOE4 is a strong genetic risk factor for Alzheimer's disease and Dementia with Lewy bodies; however, how its expression impacts pathogenic pathways in a human-relevant system is not clear. Here using human iPSC-derived cerebral organoid models, we find that APOE deletion increases α-synuclein (αSyn) accumulation accompanied with synaptic loss, reduction of GBA levels, lipid droplet accumulation and dysregulation of intracellular organelles. These phenotypes are partially rescued by exogenous apoE2 and apoE3, but not apoE4. Lipidomics analysis detects the increased fatty acid utilization and cholesterol ester accumulation in apoE-deficient cerebral organoids. Furthermore, APOE4 cerebral organoids have increased αSyn accumulation compared to those with APOE3. Carrying APOE4 also increases apoE association with Lewy bodies in postmortem brains from patients with Lewy body disease. Our findings reveal the predominant role of apoE in lipid metabolism and αSyn pathology in iPSC-derived cerebral organoids, providing mechanistic insights into how APOE4 drives the risk for synucleinopathies.


Asunto(s)
Apolipoproteínas E/metabolismo , Metabolismo de los Lípidos/fisiología , Organoides/patología , Sinucleinopatías/metabolismo , alfa-Sinucleína/metabolismo , Animales , Humanos , Células Madre Pluripotentes Inducidas , Ratones , Organoides/metabolismo , Isoformas de Proteínas/metabolismo , Sinucleinopatías/patología
2.
Alzheimers Dement ; 16(10): 1372-1383, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32827351

RESUMEN

INTRODUCTION: Cerebrovascular pathologies including cerebral amyloid angiopathy (CAA) and blood-brain barrier (BBB) dysregulation are prominent features in the majority of Alzheimer's disease (AD) cases. METHODS: We performed neuropathologic and biochemical studies on a large, neuropathologically confirmed human AD cohort (N = 469). Amounts of endothelial tight junction proteins claudin-5 (CLDN5) and occludin (OCLN), and major AD-related molecules (amyloid beta [Aß40], Aß42, tau, p-tau, and apolipoprotein E) in the temporal cortex were assessed by ELISA. RESULTS: Higher levels of soluble tau, insoluble p-tau, and apolipoprotein E (apoE) were independently correlated with lower levels of endothelial tight junction proteins CLDN5 and OCLN in AD brains. Although high Aß40 levels, APOE ε4, and male sex were predominantly associated with exacerbated CAA severity, those factors did not influence tight junction protein levels. DISCUSSION: Refining the molecular mechanisms connecting tau, Aß, and apoE with cerebrovascular pathologies is critical for greater understanding of AD pathogenesis and establishing effective therapeutic interventions for the disease.


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo/patología , Angiopatía Amiloide Cerebral , Uniones Estrechas/patología , Proteínas tau/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Apolipoproteína E4/metabolismo , Encéfalo/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Uniones Estrechas/metabolismo , Uniones Estrechas/metabolismo
3.
Hum Mol Genet ; 26(14): 2690-2700, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28444230

RESUMEN

The ε4 allele of the APOE gene encoding apolipoprotein E (apoE) is a strong genetic risk factor for aging-related cognitive decline as well as late-onset Alzheimer's disease (AD) compared to the common ε3 allele. In the central nervous system, apoE is produced primarily by astrocytes and functions in transporting lipids including cholesterol to support neuronal homeostasis and synaptic integrity. Although mouse models and corresponding primary cells have provided valuable tools for studying apoE isoform-dependent functions, recent studies have shown that human astrocytes have a distinct gene expression profile compare with rodent astrocytes. Human induced pluripotent stem cells (iPSCs) derived from individuals carrying specific gene variants or mutations provide an alternative cellular model more relevant to humans upon differentiation into specific cell types. Thus, we reprogramed human skin fibroblasts from cognitively normal individuals carrying APOE ε3/ε3 or ε4/ε4 genotype to iPSC clones and further differentiated them into neural progenitor cells and then astrocytes. We found that human iPSC-derived astrocytes secreted abundant apoE with apoE4 lipoprotein particles less lipidated compared to apoE3 particles. More importantly, human iPSC-derived astrocytes were capable of promoting neuronal survival and synaptogenesis when co-cultured with iPSC-derived neurons with APOE ε4/ε4 astrocytes less effective in supporting these neurotrophic functions than those with APOE ε3/ε3 genotype. Taken together, our findings demonstrate APOE genotype-dependent effects using human iPSC-derived astrocytes and provide novel evidence that the human iPSC-based model system is a strong tool to explore how apoE isoforms contribute to neurodegenerative diseases.


Asunto(s)
Apolipoproteína E4/genética , Astrocitos/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Alelos , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E4/metabolismo , Astrocitos/citología , Astrocitos/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Genotipo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/citología , Neuronas/metabolismo
5.
Acta Neuropathol ; 132(2): 225-234, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27179972

RESUMEN

Cerebral amyloid angiopathy (CAA) often coexists with Alzheimer's disease (AD). APOE4 is a strong genetic risk factor for both AD and CAA. Sex-dependent differences have been shown in AD as well as in cerebrovascular diseases. Therefore, we examined the effects of APOE4, sex, and pathological components on CAA in AD subjects. A total of 428 autopsied brain samples from pathologically confirmed AD cases were analyzed. CAA severity was histologically scored in inferior parietal, middle frontal, motor, superior temporal and visual cortexes. In addition, subgroups with severe CAA (n = 60) or without CAA (n = 39) were subjected to biochemical analysis of amyloid-ß (Aß) and apolipoprotein E (apoE) by ELISA in the temporal cortex. After adjusting for age, Braak neurofibrillary tangle stage and Thal amyloid phase, we found that overall CAA scores were higher in males than females. Furthermore, carrying one or more APOE4 alleles was associated with higher overall CAA scores. Biochemical analysis revealed that the levels of detergent-soluble and detergent-insoluble Aß40, and insoluble apoE were significantly elevated in individuals with severe CAA or APOE4. The ratio of Aß40/Aß42 in insoluble fractions was also increased in the presence of CAA or APOE4, although it was negatively associated with male sex. Levels of insoluble Aß40 were positively associated with those of insoluble apoE, which were strongly influenced by CAA status. Pertaining to insoluble Aß42, the levels of apoE correlated regardless of CAA status. Our results indicate that sex and APOE genotypes differentially influence the presence and severity of CAA in AD, likely by affecting interaction and aggregation of Aß40 and apoE.


Asunto(s)
Enfermedad de Alzheimer/genética , Apolipoproteína E4/genética , Encéfalo/patología , Angiopatía Amiloide Cerebral/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Angiopatía Amiloide Cerebral/patología , Femenino , Genotipo , Humanos , Masculino , Ovillos Neurofibrilares/patología , Placa Amiloide/patología , Caracteres Sexuales
6.
Nat Commun ; 11(1): 5540, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-33139712

RESUMEN

APOE4 is the strongest genetic risk factor associated with late-onset Alzheimer's disease (AD). To address the underlying mechanism, we develop cerebral organoid models using induced pluripotent stem cells (iPSCs) with APOE ε3/ε3 or ε4/ε4 genotype from individuals with either normal cognition or AD dementia. Cerebral organoids from AD patients carrying APOE ε4/ε4 show greater apoptosis and decreased synaptic integrity. While AD patient-derived cerebral organoids have increased levels of Aß and phosphorylated tau compared to healthy subject-derived cerebral organoids, APOE4 exacerbates tau pathology in both healthy subject-derived and AD patient-derived organoids. Transcriptomics analysis by RNA-sequencing reveals that cerebral organoids from AD patients are associated with an enhancement of stress granules and disrupted RNA metabolism. Importantly, isogenic conversion of APOE4 to APOE3 attenuates the APOE4-related phenotypes in cerebral organoids from AD patients. Together, our study using human iPSC-organoids recapitulates APOE4-related phenotypes and suggests APOE4-related degenerative pathways contributing to AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Organoides/metabolismo , Sinapsis/metabolismo , Enfermedad de Alzheimer/genética , Apolipoproteína E4/genética , Regulación de la Expresión Génica , Genotipo , Humanos , Organoides/patología , ARN/metabolismo , Transcriptoma
7.
Sci Transl Med ; 12(529)2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-32024798

RESUMEN

The apolipoprotein E (APOE) ε4 allele is the strongest genetic risk factor for late-onset Alzheimer's disease mainly by driving amyloid-ß pathology. Recently, APOE4 has also been found to be a genetic risk factor for Lewy body dementia (LBD), which includes dementia with Lewy bodies and Parkinson's disease dementia. How APOE4 drives risk of LBD and whether it has a direct effect on α-synuclein pathology are not clear. Here, we generated a mouse model of synucleinopathy using an adeno-associated virus gene delivery of α-synuclein in human APOE-targeted replacement mice expressing APOE2, APOE3, or APOE4. We found that APOE4, but not APOE2 or APOE3, increased α-synuclein pathology, impaired behavioral performances, worsened neuronal and synaptic loss, and increased astrogliosis at 9 months of age. Transcriptomic profiling in APOE4-expressing α-synuclein mice highlighted altered lipid and energy metabolism and synapse-related pathways. We also observed an effect of APOE4 on α-synuclein pathology in human postmortem brains with LBD and minimal amyloid pathology. Our data demonstrate a pathogenic role of APOE4 in exacerbating α-synuclein pathology independent of amyloid, providing mechanistic insights into how APOE4 increases the risk of LBD.


Asunto(s)
Apolipoproteína E4 , Enfermedad por Cuerpos de Lewy/genética , Sinucleinopatías , alfa-Sinucleína , Péptidos beta-Amiloides , Animales , Apolipoproteína E4/genética , Ratones , Ratones Noqueados para ApoE , Sinucleinopatías/genética
8.
Cell Metab ; 30(6): 1131-1140.e7, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31564441

RESUMEN

The identification of molecular targets and pharmacodynamic markers for Parkinson's disease (PD) will empower more effective clinical management and experimental therapies. Miro1 is localized on the mitochondrial surface and mediates mitochondrial motility. Miro1 is removed from depolarized mitochondria to facilitate their clearance via mitophagy. Here, we explore the clinical utility of Miro1 for detecting PD and for gauging potential treatments. We measure the Miro1 response to mitochondrial depolarization using biochemical assays in skin fibroblasts from a broad spectrum of PD patients and discover that more than 94% of the patients' fibroblast cell lines fail to remove Miro1 following depolarization. We identify a small molecule that can repair this defect of Miro1 in PD fibroblasts. Treating patient-derived neurons and fly models with this compound rescues the locomotor deficits and dopaminergic neurodegeneration. Our results indicate that tracking this Miro1 marker and engaging in Miro1-based therapies could open new avenues to personalized medicine.


Asunto(s)
Antiparkinsonianos/farmacología , Proteínas de Drosophila , Proteínas Mitocondriales , Degeneración Nerviosa/tratamiento farmacológico , Neuronas/efectos de los fármacos , Enfermedad de Parkinson , Proteínas de Unión al GTP rho , Adulto , Anciano , Animales , Antiparkinsonianos/uso terapéutico , Biomarcadores/metabolismo , Drosophila , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/metabolismo , Femenino , Fibroblastos , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/metabolismo , Neuronas/patología , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteínas de Unión al GTP rho/metabolismo
10.
Mol Neurodegener ; 10: 46, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26373282

RESUMEN

BACKGROUND: Pallido-ponto-nigral degeneration (PPND), a major subtype of frontotemporal dementia with parkinsonism related to chromosome 17 (FTDP-17), is a progressive and terminal neurodegenerative disease caused by c.837 T > G mutation in the MAPT gene encoding microtubule-associated protein tau (rs63750756; N279K). This MAPT mutation induces alternative splicing of exon 10, resulting in a modification of microtubule-binding region of tau. Although mutations in the MAPT gene have been linked to multiple tauopathies including Alzheimer's disease, frontotemporal dementia and progressive supranuclear palsy, knowledge regarding how tau N279K mutation causes PPND/FTDP-17 is limited. RESULTS: We investigated the underlying disease mechanism associated with the N279K tau mutation using PPND/FTDP-17 patient-derived induced pluripotent stem cells (iPSCs) and autopsy brains. In iPSC-derived neural stem cells (NSCs), the N279K tau mutation induced an increased ratio of 4-repeat to 3-repeat tau and accumulation of stress granules indicating elevated cellular stress. More significant, NSCs derived from patients with the N279K tau mutation displayed impaired endocytic trafficking as evidenced by accumulation of endosomes and exosomes, and a reduction of lysosomes. Since there were no significant differences in cellular stress and distribution of subcellular organelles between control and N279K skin fibroblasts, N279K-related vesicle trafficking defects are likely specific to the neuronal lineage. Consistently, the levels of intracellular/luminal vesicle and exosome marker flotillin-1 were significantly increased in frontal and temporal cortices of PPND/FTDP-17 patients with the N279K tau mutation, events that were not seen in the occipital cortex which is the most spared cortical region in the patients. CONCLUSION: Together, our results demonstrate that alterations of intracellular vesicle trafficking in NSCs/neurons likely contribute to neurodegeneration as an important disease mechanism underlying the N279K tau mutation in PPND/FTDP-17.


Asunto(s)
Demencia Frontotemporal/genética , Células Madre Pluripotentes Inducidas/metabolismo , Mutación Missense , Mutación Puntual , Transporte de Proteínas , Vesículas Transportadoras/fisiología , Proteínas tau/genética , Sitios de Unión/genética , Células Cultivadas , Cuerpos Embrioides , Endosomas/fisiología , Exones/genética , Exosomas/fisiología , Femenino , Lóbulo Frontal/metabolismo , Lóbulo Frontal/patología , Demencia Frontotemporal/metabolismo , Demencia Frontotemporal/patología , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/citología , Lisosomas/fisiología , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Persona de Mediana Edad , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Lóbulo Occipital/metabolismo , Lóbulo Occipital/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Lóbulo Temporal/metabolismo , Lóbulo Temporal/patología
11.
Nat Neurosci ; 18(8): 1175-82, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26192745

RESUMEN

Increasing evidence suggests that defective RNA processing contributes to the development of amyotrophic lateral sclerosis (ALS). This may be especially true for ALS caused by a repeat expansion in C9orf72 (c9ALS), in which the accumulation of RNA foci and dipeptide-repeat proteins are expected to modify RNA metabolism. We report extensive alternative splicing (AS) and alternative polyadenylation (APA) defects in the cerebellum of c9ALS subjects (8,224 AS and 1,437 APA), including changes in ALS-associated genes (for example, ATXN2 and FUS), and in subjects with sporadic ALS (sALS; 2,229 AS and 716 APA). Furthermore, heterogeneous nuclear ribonucleoprotein H (hnRNPH) and other RNA-binding proteins are predicted to be potential regulators of cassette exon AS events in both c9ALS and sALS. Co-expression and gene-association network analyses of gene expression and AS data revealed divergent pathways associated with c9ALS and sALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Cerebelo/metabolismo , Lóbulo Frontal/metabolismo , Regulación de la Expresión Génica/genética , Proteínas/genética , ARN/metabolismo , Transcriptoma/genética , Adulto , Anciano , Empalme Alternativo , Proteína C9orf72 , Estudios de Asociación Genética , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/metabolismo , Humanos , Persona de Mediana Edad , Poliadenilación/genética , Análisis de Secuencia de ARN
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda