Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Neurobiol Dis ; 153: 105303, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33631273

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, robust microgliosis, neuroinflammation, and neuronal loss. Genome-wide association studies recently highlighted a prominent role for microglia in late-onset AD (LOAD). Specifically, inositol polyphosphate-5-phosphatase (INPP5D), also known as SHIP1, is selectively expressed in brain microglia and has been reported to be associated with LOAD. Although INPP5D is likely a crucial player in AD pathophysiology, its role in disease onset and progression remains unclear. We performed differential gene expression analysis to investigate INPP5D expression in AD and its association with plaque density and microglial markers using transcriptomic (RNA-Seq) data from the Accelerating Medicines Partnership for Alzheimer's Disease (AMP-AD) cohort. We also performed quantitative real-time PCR, immunoblotting, and immunofluorescence assays to assess INPP5D expression in the 5xFAD amyloid mouse model. Differential gene expression analysis found that INPP5D expression was upregulated in LOAD and positively correlated with amyloid plaque density. In addition, in 5xFAD mice, Inpp5d expression increased as the disease progressed, and selectively in plaque-associated microglia. Increased Inpp5d expression levels in 5xFAD mice were abolished entirely by depleting microglia with the colony-stimulating factor receptor-1 antagonist PLX5622. Our findings show that INPP5D expression increases as AD progresses, predominantly in plaque-associated microglia. Importantly, we provide the first evidence that increased INPP5D expression might be a risk factor in AD, highlighting INPP5D as a potential therapeutic target. Moreover, we have shown that the 5xFAD mouse model is appropriate for studying INPP5D in AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Microglía/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/genética , Placa Amiloide/genética , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/metabolismo , Placa Amiloide/metabolismo , ARN Mensajero/metabolismo , RNA-Seq
2.
Neurobiol Dis ; 142: 104956, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32479996

RESUMEN

Alzheimer's disease (AD) is a prominent neurodegenerative disorder characterized by deposition of ß-amyloid (Aß)-containing extracellular plaques, accompanied by a microglial-mediated inflammatory response, that leads to cognitive decline. Microglia perform many disease-modifying functions such as phagocytosis of plaques, plaque compaction, and modulation of inflammation through the secretion of cytokines. Microglia are reliant upon colony-stimulating factor receptor-1 (CSF1R) activation for survival. In AD mouse models, chronic targeted depletion of microglia via CSF1R antagonism attenuates plaque formation in early disease but fails to alter plaque burden in late disease. It is unclear if acute depletion of microglia during the peak period of plaque deposition will alter disease pathogenesis, and if so, whether these effects are reversible upon microglial repopulation. To test this, we administered the CSF1R antagonist PLX5622 to the 5XFAD mouse model of AD at four months of age for approximately one month. In a subset of mice, the drug treatment was discontinued, and the mice were fed a control diet for an additional month. We evaluated plaque burden and composition, microgliosis, inflammatory marker expression, and neuritic dystrophy. In 5XFAD animals, CSF1R blockade for 28 days depleted microglia across brain regions by over 50%, suppressed microgliosis, and reduced plaque burden. In microglial-depleted AD animals, neuritic dystrophy was enhanced, and increased diffuse-like plaques and fewer compact-like plaques were observed. Removal of PLX5622 elicited microglial repopulation and subsequent plaque remodeling, resulting in more compact plaques predominating microglia-repopulated regions. We found that microglia limit diffuse plaques by maintaining compact-like plaque properties, thereby blocking the progression of neuritic dystrophy. Microglial repopulation reverses these effects. Collectively, we show that microglia are neuroprotective through maintenance of plaque compaction and morphologies during peak disease progression.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Microglía/metabolismo , Placa Amiloide/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Ratones Transgénicos , Microglía/patología , Compuestos Orgánicos/farmacología , Placa Amiloide/patología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores
3.
J Neuroinflammation ; 15(1): 43, 2018 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-29448961

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is a highly prevalent neurodegenerative disorder characterized by pathological hallmarks of beta-amyloid plaque deposits, tau pathology, inflammation, and cognitive decline. Treatment remains a clinical obstacle due to lack of effective therapeutics. Agonists targeting nuclear receptors, such as bexarotene, reversed cognitive deficits regardless of treatment duration and age in murine models of AD. While bexarotene demonstrated marked efficacy in decreasing plaque levels following short-term treatment, prolonged treatment did not modulate plaque burden. This suggested that plaques might reform in mice treated chronically with bexarotene and that cessation of bexarotene treatment before plaques reform might alter amyloid pathology, inflammation, and cognition in AD mice. METHODS: We utilized one-year-old APP/PS1 mice that were divided into two groups. We treated one group of mice for 2 weeks with bexarotene. The other group of mice was treated for 2 weeks with bexarotene followed by withdrawal of drug treatment for an additional 2 weeks. Cognition was evaluated using the novel-object recognition test either at the end of bexarotene treatment or the end of the withdrawal period. We then analyzed amyloid pathology and microgliosis at the conclusion of the study in both groups. RESULTS: Bexarotene treatment enhanced cognition in APP/PS1 mice similar to previous findings. Strikingly, we observed sustained cognitive improvements in mice in which bexarotene treatment was discontinued for 2 weeks. We observed a sustained reduction in microgliosis and plaque burden following drug withdrawal exclusively in the hippocampus. CONCLUSIONS: Our findings demonstrate that bexarotene selectively modifies aspects of neuroinflammation in a region-specific manner to reverse hippocampal-dependent cognitive deficits in AD mice and may provide insight to inform future studies with nuclear receptor agonists.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Bexaroteno/uso terapéutico , Trastornos del Conocimiento/tratamiento farmacológico , Modelos Animales de Enfermedad , Placa Amiloide/tratamiento farmacológico , Receptores X Retinoide/agonistas , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/análisis , Animales , Animales Recién Nacidos , Bexaroteno/farmacología , Células Cultivadas , Cognición/efectos de los fármacos , Cognición/fisiología , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/análisis , Placa Amiloide/metabolismo , Placa Amiloide/patología , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores X Retinoide/antagonistas & inhibidores , Receptores X Retinoide/metabolismo
4.
J Neurosci ; 35(24): 9173-81, 2015 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-26085639

RESUMEN

Alzheimer's disease (AD) is a highly prevalent disorder for which there are no effective therapies. Accumulation of amyloid ß (Aß) peptides in the brain is associated with impaired cognition and memory, pronounced inflammatory dysregulation, and subsequent amyloid plaque deposition. Thus, drugs that promote the clearance of Aß peptides and resolution of inflammation may represent viable therapeutic approaches. Agonists of nuclear receptors LXR:RXR and PPAR:RXR act to ameliorate AD-related cognitive impairment and amyloid accumulation in murine models of AD. The use of an agonist to the nuclear receptor RXR, bexarotene, as monotherapy against AD, presents potential challenges due to the metabolic perturbations it induces in the periphery, most prominently hypertriglyceridemia. We report that the ω-3 fatty acid docosahexaenoic acid (DHA), in combination with bexarotene, enhances LXR:RXR target gene expression of Abca1 and ApoE, reduces soluble forms of Aß, and abrogates release of pro-inflammatory cytokines and mediators both in vitro and in a mouse model of AD. Moreover, DHA abrogates bexarotene-induced hypertriglyceridemia in vivo. Importantly, dual therapy promotes reductions in AD pathology and resultant amelioration of cognitive deficits. While monotherapy with either bexarotene or DHA resulted in modest effects in vitro and in vivo, combined treatment with both agents produced a significant additive benefit on associated AD-related phenotypes, suggesting that targeted combinatorial agents may be beneficial over single agents alone in treating AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Modelos Animales de Enfermedad , Ácidos Grasos Omega-3/administración & dosificación , Receptores Citoplasmáticos y Nucleares/agonistas , Tetrahidronaftalenos/administración & dosificación , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Bexaroteno , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Ácidos Docosahexaenoicos/administración & dosificación , Quimioterapia Combinada , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Citoplasmáticos y Nucleares/metabolismo
5.
J Biol Chem ; 290(35): 21591-602, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26163517

RESUMEN

Alzheimer disease (AD) is characterized by the extracellular accumulation of amyloid ß (Aß), which is accompanied by a robust inflammatory response in the brain. Both of these pathogenic processes are regulated by nuclear receptors, including the liver X receptors (LXRs) and peroxisome-proliferator receptor γ (PPARγ). Agonists of LXRs have been demonstrated previously to reduce Aß levels and improve cognitive deficits in AD mouse models by inducing the transcription and lipidation of apolipoprotein E (apoE). Agonists targeting PPARγ reduce the microglial expression of proinflammatory genes and have also been shown to modulate apoE expression. Here we investigate whether a combination therapy with both LXR and PPARγ agonists results in increased benefits in an AD mouse model. We found that the LXR agonist GW3965 and the PPARγ agonist pioglitazone were individually able to increase the levels of apoE and related genes, decrease the expression of proinflammatory genes, and facilitate Aß decreases in the hippocampus. Combined treatment with both agonists provoked a further increase in the expression of apoE and a decrease in the soluble and deposited forms of Aß. The decrease in plaques was associated with increased colocalization between microglia and plaques. In addition, the PPARγ agonist in the combined treatment paradigm was able to counteract the elevation in plasma triglycerides that is a side effect of LXR agonist treatment. These results suggest that combined LXR/PPARγ agonist treatment merits further investigation for the treatment of AD.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Conducta Animal , Receptores Nucleares Huérfanos/agonistas , PPAR gamma/agonistas , Presenilina-1/metabolismo , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Apolipoproteínas E/metabolismo , Conducta Animal/efectos de los fármacos , Benzoatos/farmacología , Bencilaminas/farmacología , Biomarcadores/metabolismo , Trastornos del Conocimiento/complicaciones , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Mediadores de Inflamación/metabolismo , Receptores X del Hígado , Masculino , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Receptores Nucleares Huérfanos/metabolismo , PPAR gamma/metabolismo , Tamaño de la Partícula , Pioglitazona , Placa Amiloide/metabolismo , Placa Amiloide/patología , Proteolisis/efectos de los fármacos , Tiazolidinedionas/farmacología , Transcripción Genética/efectos de los fármacos , Triglicéridos/sangre
6.
bioRxiv ; 2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-38014106

RESUMEN

Microgliosis and neuroinflammation are prominent features of Alzheimer's disease (AD). Disease-responsive microglia meet their increased energy demand by reprogramming metabolism, specifically, switching to favor glycolysis over oxidative phosphorylation. Thus, targeting of microglial immunometabolism might be of therapeutic benefit for treating AD, providing novel and often well understood immune pathways and their newly recognized actions in AD. We report that in the brains of 5xFAD mice and postmortem brains of AD patients, we found a significant increase in the levels of Hexokinase 2 (HK2), an enzyme that supports inflammatory responses by rapidly increasing glycolysis. Moreover, binding of HK2 to mitochondria has been reported to regulate inflammation by preventing mitochondrial dysfunction and NLRP3 inflammasome activation, suggesting that its inflammatory role extends beyond its glycolytic activity. Here we report, that HK2 antagonism selectively affects microglial phenotypes and disease progression in a gene-dose dependent manner. Paradoxically, complete loss of HK2 fails to improve AD progression by exacerbating inflammasome activity while its haploinsufficiency results in reduced pathology and improved cognition in the 5XFAD mice. We propose that the partial antagonism of HK2, is effective in slowed disease progression and inflammation through a non-metabolic mechanism associated with the modulation of NFKß signaling, through its cytosolic target IKBα. The complete loss of HK2 affects additional inflammatory mechanisms associated to mitochondrial dysfunction.

7.
Genome Med ; 14(1): 17, 2022 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-35180881

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is characterized by robust microgliosis and phenotypic changes that accompany disease pathogenesis. Accumulating evidence from genetic studies suggests the importance of phospholipase C γ 2 (PLCG2) in late-onset AD (LOAD) pathophysiology. However, the role of PLCG2 in AD is still poorly understood. METHODS: Using bulk RNA-Seq (N=1249) data from the Accelerating Medicines Partnership-Alzheimer's Disease Consortium (AMP-AD), we investigated whether PLCG2 expression increased in the brains of LOAD patients. We also evaluated the relationship between PLCG2 expression levels, amyloid plaque density, and expression levels of microglia specific markers (AIF1 and TMEM119). Finally, we investigated the longitudinal changes of PLCG2 expression in the 5xFAD mouse model of AD. To further understand the role of PLCG2 in different signaling pathways, differential gene expression and co-expression network analyses were performed using bulk RNA-Seq and microglial single-cell RNA-Seq data. To substantiate the human analyses, we performed differential gene expression analysis on wild-type (WT) and inactivated Plcg2 mice and used immunostaining to determine if the differentially expressed genes/pathways were altered by microglial cell coverage or morphology. RESULTS: We observed significant upregulation of PLCG2 expression in three brain regions of LOAD patients and significant positive correlation of PLCG2 expression with amyloid plaque density. These findings in the human brain were validated in the 5xFAD amyloid mouse model, which showed disease progression-dependent increases in Plcg2 expression associated with amyloid pathology. Of note, increased Plcg2 expression levels in 5xFAD mice were abolished by reducing microglia. Furthermore, using bulk RNA-Seq data, we performed differential expression analysis by comparing cognitively normal older adults (CN) with 75th percentile (high) and 25th percentile (low) PLCG2 gene expression levels to identify pathways related to inflammation and the inflammatory response. The findings in the human brain were validated by differential expression analyses between WT and plcg2 inactivated mice. PLCG2 co-expression network analysis of microglial single-cell RNA-Seq data identified pathways related to the inflammatory response including regulation of I-kappaB/NF-kappa B signaling and response to lipopolysaccharide. CONCLUSIONS: Our results provide further evidence that PLCG2 plays an important role in AD pathophysiology and may be a potential target for microglia-targeted AD therapies.


Asunto(s)
Enfermedad de Alzheimer , Placa Amiloide , Anciano , Enfermedad de Alzheimer/patología , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Microglía/metabolismo , Fosfolipasa C gamma/genética , Fosfolipasa C gamma/metabolismo , Placa Amiloide/genética , Placa Amiloide/metabolismo , Placa Amiloide/patología
8.
Sci Transl Med ; 14(637): eabl7634, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35320002

RESUMEN

Increased dietary intake of niacin has been correlated with reduced risk of Alzheimer's disease (AD). Niacin serves as a high-affinity ligand for the receptor HCAR2 (GPR109A). In the brain, HCAR2 is expressed selectively by microglia and is robustly induced by amyloid pathology in AD. The genetic inactivation of Hcar2 in 5xFAD mice, a model of AD, results in impairment of the microglial response to amyloid deposition, including deficits in gene expression, proliferation, envelopment of amyloid plaques, and uptake of amyloid-ß (Aß), ultimately leading to exacerbation of amyloid burden, neuronal loss, and cognitive deficits. In contrast, activation of HCAR2 with an FDA-approved formulation of niacin (Niaspan) in 5xFAD mice leads to reduced plaque burden and neuronal dystrophy, attenuation of neuronal loss, and rescue of working memory deficits. These data provide direct evidence that HCAR2 is required for an efficient and neuroprotective response of microglia to amyloid pathology. Administration of Niaspan potentiates the HCAR2-mediated microglial protective response and consequently attenuates amyloid-induced pathology, suggesting that its use may be a promising therapeutic approach to AD that specifically targets the neuroimmune response.


Asunto(s)
Enfermedad de Alzheimer , Niacina , Receptores Acoplados a Proteínas G , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ratones , Ratones Transgénicos , Microglía/metabolismo , Niacina/farmacología , Placa Amiloide/patología , Receptores Acoplados a Proteínas G/metabolismo
9.
Cells ; 10(4)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33924200

RESUMEN

Microglia are the resident immune cells of the brain, deriving from yolk sac progenitors that populate the brain parenchyma during development. During development and homeostasis, microglia play critical roles in synaptogenesis and synaptic plasticity, in addition to their primary role as immune sentinels. In aging and neurodegenerative diseases generally, and Alzheimer's disease (AD) specifically, microglial function is altered in ways that significantly diverge from their homeostatic state, inducing a more detrimental inflammatory environment. In this review, we discuss the receptors, signaling, regulation and gene expression patterns of microglia that mediate their phenotype and function contributing to the inflammatory milieu of the AD brain, as well as strategies that target microglia to ameliorate the onset, progression and symptoms of AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Desarrollo Embrionario , Homeostasis , Microglía/metabolismo , Envejecimiento/metabolismo , Animales , Humanos , Microglía/inmunología , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/metabolismo
10.
Mol Neurodegener ; 13(1): 29, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29859094

RESUMEN

BACKGROUND: The R47H variant of Triggering Receptor Expressed on Myeloid cells 2 (TREM2) confers greatly increased risk for Alzheimer's disease (AD), reflective of a central role for myeloid cells in neurodegeneration. Understanding how this variant confers AD risk promises to provide important insights into how myeloid cells contribute to AD pathogenesis and progression. METHODS: In order to investigate this mechanism, CRISPR/Cas9 was used to generate a mouse model of AD harboring one copy of the single nucleotide polymorphism (SNP) encoding the R47H variant in murine Trem2. TREM2 expression, myeloid cell responses to amyloid deposition, plaque burden, and neuritic dystrophy were assessed at 4 months of age. RESULTS: AD mice heterozygous for the Trem2 R47H allele exhibited reduced total Trem2 mRNA expression, reduced TREM2 expression around plaques, and reduced association of myeloid cells with plaques. These results were comparable to AD mice lacking one copy of Trem2. AD mice heterozygous for the Trem2 R47H allele also showed reduced myeloid cell responses to amyloid deposition, including a reduction in proliferation and a reduction in CD45 expression around plaques. Expression of the Trem2 R47H variant also reduced dense core plaque number but increased plaque-associated neuritic dystrophy. CONCLUSIONS: These data suggest that the AD-associated TREM2 R47H variant increases risk for AD by conferring a loss of TREM2 function and enhancing neuritic dystrophy around plaques.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo/patología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Animales , Glicoproteínas de Membrana/genética , Ratones , Fenotipo , Polimorfismo de Nucleótido Simple , Receptores Inmunológicos/genética
11.
Bio Protoc ; 6(8)2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-28516120

RESUMEN

This protocol details beta-amyloid (Aß) extraction from transgenic murine brain homogenates. Specifically, mechanical homogenization of brain tissue and sequential extraction of both soluble and insoluble proteins are detailed. DEA extracts soluble proteins, such as Aß isoforms and APP. Formic acid enables extraction of insoluble protein aggregates, such as Aß isoforms associated with plaques. This procedure produces soluble and insoluble extracts that are amenable to analysis of Aß species via western blotting and/or enzyme-linked immunosorbent assays (ELISAs), and these results help assess amyloidogenic burden in animals.

12.
J Neuroimmune Pharmacol ; 11(1): 61-72, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26175148

RESUMEN

Alzheimer's disease (AD) is characterized by impaired clearance of amyloid beta (Aß) peptides, leading to the accumulation of Aß in the brain and subsequent neurodegeneration and cognitive impairment. ApoE plays a critical role in the proteolytic degradation of soluble forms of Aß. This effect is dependent upon lipidation of ApoE by ABCA1-mediated transfer of phospholipids and cholesterol. ApoE and ABCA1 are induced by the action of the RXR agonist, bexarotene. We have previously shown that bexarotene reduces Aß levels in AD mouse models and we have hypothesized that this effect requires ABCA1-mediated lipidation of ApoE. To test this hypothesis, we crossed ABCA1-deficient (ABCA1 KO) mice with the APP/PS1 model of AD. Aged ABCA1 WT and ABCA1 KO APP/PS1 mice were treated for 7 days with vehicle or bexarotene (100 mg/kg/day). Bexarotene reduced levels of soluble Aß 1-40 and 1-42 in the hippocampus of ABCA1 WT but not ABCA1 KO APP/PS1 mice. In contrast, insoluble levels of Aß, and plaque loads were unaffected by bexarotene in this study. ABCA1 KO mice had increased levels of inflammation compared with ABCA1 WT mice. Bexarotene also increased most inflammatory gene markers evaluated. The effect of bexarotene on microglial inflammatory profiles, however, was independent of ABCA1 genotype. Importantly, bexarotene ameliorated deficits in novel object recognition in ABCA1 WT but not ABCA1 KO APP/PS1 mice. These data indicate that ABCA1-induced lipidation of ApoE is necessary for the ability of bexarotene to clear hippocampal soluble Aß and ameliorate cognitive deficits.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E/metabolismo , Hipocampo/metabolismo , Tetrahidronaftalenos/farmacología , Precursor de Proteína beta-Amiloide/genética , Animales , Bexaroteno , Western Blotting , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Hipocampo/efectos de los fármacos , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Presenilina-1/genética
13.
Science ; 335(6075): 1503-6, 2012 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-22323736

RESUMEN

Alzheimer's disease (AD) is associated with impaired clearance of ß-amyloid (Aß) from the brain, a process normally facilitated by apolipoprotein E (apoE). ApoE expression is transcriptionally induced through the action of the nuclear receptors peroxisome proliferator-activated receptor gamma and liver X receptors in coordination with retinoid X receptors (RXRs). Oral administration of the RXR agonist bexarotene to a mouse model of AD resulted in enhanced clearance of soluble Aß within hours in an apoE-dependent manner. Aß plaque area was reduced more than 50% within just 72 hours. Furthermore, bexarotene stimulated the rapid reversal of cognitive, social, and olfactory deficits and improved neural circuit function. Thus, RXR activation stimulates physiological Aß clearance mechanisms, resulting in the rapid reversal of a broad range of Aß-induced deficits.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E/metabolismo , Encéfalo/metabolismo , Tetrahidronaftalenos/farmacología , Tetrahidronaftalenos/uso terapéutico , Amiloidosis/tratamiento farmacológico , Amiloidosis/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Conducta Animal/efectos de los fármacos , Bexaroteno , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Líquido Extracelular/efectos de los fármacos , Líquido Extracelular/metabolismo , Receptores X del Hígado , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/metabolismo , Terapia Molecular Dirigida , Odorantes , Vías Olfatorias/efectos de los fármacos , Vías Olfatorias/fisiología , Receptores Nucleares Huérfanos/metabolismo , PPAR gamma/metabolismo , Fagocitosis , Placa Amiloide/tratamiento farmacológico , Receptores X Retinoide/agonistas , Receptores X Retinoide/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA