Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Brain ; 146(6): 2268-2274, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37127299

ABSTRACT

The risk of developing Alzheimer's disease is mediated by a combination of genetics and environmental factors, such as stress, sleep abnormalities and traumatic brain injury. Women are at a higher risk of developing Alzheimer's disease than men, even when controlling for differences in lifespan. Women are also more likely to report high levels of stress than men. Sex differences in response to stress may play a role in the increased risk of Alzheimer's disease in women. In this study, we use in vivo microdialysis to measure levels of Aß in response to acute stress in male and female mice. We show that Aß levels are altered differently between female and male mice (APP/PS1 and wild-type) in response to stress, with females showing significantly increased levels of Aß while most males do not show a significant change. This response is mediated through ß-arrestin involvement in Corticotrophin Releasing Factor receptor signalling pathway differences in male and female mice as male mice lacking ß-arrestin show increase in Aß in response to stress similar to females.


Subject(s)
Alzheimer Disease , Mice , Female , Male , Animals , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Mice, Transgenic , Disease Models, Animal , Amyloid beta-Peptides/metabolism , beta-Arrestins/metabolism , Presenilin-1/metabolism
2.
J Neurochem ; 156(5): 658-673, 2021 03.
Article in English | MEDLINE | ID: mdl-33278025

ABSTRACT

Amyloid-ß (Aß) peptide aggregation into soluble oligomers and insoluble plaques is a precipitating event in the pathogenesis of Alzheimer's disease (AD). Given that synaptic activity can regulate Aß generation, we postulated that 5HT2A -Rs may regulate Aß as well. We treated APP/PS1 transgenic mice with the selective 5HT2A inverse agonists M100907 or Pimavanserin systemically and measured brain interstitial fluid (ISF) Aß levels in real-time using in vivo microdialysis. Both compounds reduced ISF Aß levels by almost 50% within hours, but had no effect on Aß levels in 5HT2A -R knock-out mice. The Aß-lowering effects of Pimavanserin were blocked by extracellular-regulated kinase (ERK) and NMDA receptor inhibitors. Chronic administration of Pimavanserin by subcutaneous osmotic pump to aged APP/PS1 mice significantly reduced CSF Aß levels and Aß pathology and improved cognitive function in these mice. Pimavanserin is FDA-approved to treat Parkinson's disease psychosis, and also has been shown to reduce psychosis in a variety of other dementia subtypes including Alzheimer's disease. These data demonstrate that Pimavanserin may have disease-modifying benefits in addition to its efficacy against neuropsychiatric symptoms of Alzheimer's disease. Read the Editorial Highlight for this article on page 560.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Drug Inverse Agonism , Piperidines/therapeutic use , Receptor, Serotonin, 5-HT2A/metabolism , Serotonin 5-HT2 Receptor Agonists/therapeutic use , Urea/analogs & derivatives , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/biosynthesis , Animals , Female , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred C3H , Mice, Transgenic , Piperidines/pharmacology , Serotonin 5-HT2 Receptor Agonists/pharmacology , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Serotonin 5-HT2 Receptor Antagonists/therapeutic use , Urea/pharmacology , Urea/therapeutic use
3.
Neurology ; 95(19): e2666-e2674, 2020 11 10.
Article in English | MEDLINE | ID: mdl-32913022

ABSTRACT

BACKGROUND: Several neurotransmitter receptors activate signaling pathways that alter processing of the amyloid precursor protein (APP) into ß-amyloid (Aß). Serotonin signaling through a subset of serotonin receptors suppresses Aß generation. We proposed that escitalopram, the most specific selective serotonin reuptake inhibitor (SSRI) that inhibits the serotonin transporter SERT, would suppress Aß levels in mice. OBJECTIVES: We hypothesized that acute treatment with escitalopram would reduce Aß generation, which would be reflected chronically with a significant reduction in Aß plaque load. METHODS: We performed in vivo microdialysis and in vivo 2-photon imaging to assess changes in brain interstitial fluid (ISF) Aß and Aß plaque size over time, respectively, in the APP/presenilin 1 mouse model of Alzheimer disease treated with vehicle or escitalopram. We also chronically treated mice with escitalopram to determine the effect on plaques histologically. RESULTS: Escitalopram acutely reduced ISF Aß by 25% by increasing α-secretase cleavage of APP. Chronic administration of escitalopram significantly reduced plaque load by 28% and 34% at 2.5 and 5 mg/d, respectively. Escitalopram at 5 mg/kg did not remove existing plaques, but completely arrested individual plaque growth over time. CONCLUSIONS: Escitalopram significantly reduced Aß in mice, similar to previous findings in humans treated with acute dosing of an SSRI.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/drug effects , Brain/drug effects , Citalopram/pharmacology , Peptide Fragments/drug effects , Plaque, Amyloid/pathology , Selective Serotonin Reuptake Inhibitors/pharmacology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Extracellular Fluid , Intravital Microscopy , Mice , Microdialysis , Microscopy, Fluorescence, Multiphoton , Peptide Fragments/metabolism , Plaque, Amyloid/metabolism , Presenilin-1/genetics
4.
J Exp Med ; 215(5): 1349-1364, 2018 05 07.
Article in English | MEDLINE | ID: mdl-29626114

ABSTRACT

Drug development for Alzheimer's disease has endeavored to lower amyloid ß (Aß) by either blocking production or promoting clearance. The benefit of combining these approaches has been examined in mouse models and shown to improve pathological measures of disease over single treatment; however, the impact on cellular and cognitive functions affected by Aß has not been tested. We used a controllable APP transgenic mouse model to test whether combining genetic suppression of Aß production with passive anti-Aß immunization improved functional outcomes over either treatment alone. Compared with behavior before treatment, arresting further Aß production (but not passive immunization) was sufficient to stop further decline in spatial learning, working memory, and associative memory, whereas combination treatment reversed each of these impairments. Cognitive improvement coincided with resolution of neuritic dystrophy, restoration of synaptic density surrounding deposits, and reduction of hyperactive mammalian target of rapamycin signaling. Computational modeling corroborated by in vivo microdialysis pointed to the reduction of soluble/exchangeable Aß as the primary driver of cognitive recovery.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/antagonists & inhibitors , Cognition , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Axons/metabolism , Behavior, Animal , Biomarkers/metabolism , Drug Therapy, Combination , Immunization, Passive , Lysosomes/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Solubility , Synapses/metabolism , Transgenes
5.
Mol Neurodegener ; 11(1): 45, 2016 06 18.
Article in English | MEDLINE | ID: mdl-27315796

ABSTRACT

BACKGROUND: The aggregation of amyloid-ß (Aß) into insoluble plaques is a hallmark pathology of Alzheimer's disease (AD). Previous work has shown increasing serotonin levels with selective serotonin re-uptake inhibitor (SSRI) compounds reduces Aß in the brain interstitial fluid (ISF) in a mouse model of AD and in the cerebrospinal fluid of humans. We investigated which serotonin receptor (5-HTR) subtypes and downstream effectors were responsible for this reduction. RESULTS: Agonists of 5-HT4R, 5-HT6R, and 5-HT7R significantly reduced ISF Aß, but agonists of other receptor subtypes did not. Additionally, inhibition of Protein Kinase A (PKA) blocked the effects of citalopram, an SSRI, on ISF Aß levels. Serotonin signaling does not appear to change gene expression to reduce Aß levels in acute timeframes, but likely acts within the cytoplasm to increase α-secretase enzymatic activity. Broad pharmacological inhibition of putative α-secretases increased ISF Aß and blocked the effects of citalopram. CONCLUSIONS: In total, these studies map the major signaling components linking serotonin receptors to suppression of brain ISF Aß. These results suggest the reduction in ISF Aß is mediated by a select group of 5-HTRs and open future avenues for targeted therapy of AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloidogenic Proteins/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Plaque, Amyloid/metabolism , Receptors, Serotonin/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Disease Models, Animal , Mice , Microdialysis/methods , Plaque, Amyloid/pathology
6.
J Exp Med ; 213(5): 677-85, 2016 05 02.
Article in English | MEDLINE | ID: mdl-27069115

ABSTRACT

Findings from genetic, animal model, and human studies support the observation that accumulation of the ß-amyloid (Aß) peptide in the brain plays a central role in the pathogenic cascade of Alzheimer's disease (AD). Human studies suggest that one key factor leading to accumulation is a defect in brain Aß clearance. We have developed a novel microimmunoelectrode (MIE) to study the kinetics of Aß clearance using an electrochemical approach. This is the first study using MIEs in vivo to measure rapid changes in Aß levels in the brains of living mice. Extracellular, interstitial fluid (ISF) Aß levels were measured in the hippocampus of APP/PS1 mice. Baseline levels of Aß40 in the ISF are relatively stable and begin to decline within minutes of blocking Aß production with a γ-secretase inhibitor. Pretreatment with a P-glycoprotein inhibitor, which blocks blood-brain barrier transport of Aß, resulted in significant prolongation of Aß40 half-life, but only in the latter phase of Aß clearance from the ISF.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Extracellular Fluid/metabolism , Hippocampus/metabolism , Peptide Fragments/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Animals , Disease Models, Animal , Electrodes , Humans , Mice , Mice, Mutant Strains , Peptide Fragments/genetics
7.
Org Lett ; 16(10): 2676-9, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24785543

ABSTRACT

The synthesis of C-3-labeled phenylthio sialic acid derivatives and an investigation of stereoselectivity in elimination reactions for the synthesis of 2,3-dehydro derivatives (glycals) is described. The experimental results are consistent with the existence of a conformational change and may be indicative of the intermediacy of an all-axial oxacarbenium ion.


Subject(s)
Sialic Acids/chemical synthesis , Sulfur Compounds/chemical synthesis , Molecular Structure , Sialic Acids/chemistry , Stereoisomerism , Sulfur Compounds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...