Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Brain ; 141(8): 2457-2474, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29945247

ABSTRACT

Down syndrome, caused by trisomy of chromosome 21, is the single most common risk factor for early-onset Alzheimer's disease. Worldwide approximately 6 million people have Down syndrome, and all these individuals will develop the hallmark amyloid plaques and neurofibrillary tangles of Alzheimer's disease by the age of 40 and the vast majority will go on to develop dementia. Triplication of APP, a gene on chromosome 21, is sufficient to cause early-onset Alzheimer's disease in the absence of Down syndrome. However, whether triplication of other chromosome 21 genes influences disease pathogenesis in the context of Down syndrome is unclear. Here we show, in a mouse model, that triplication of chromosome 21 genes other than APP increases amyloid-ß aggregation, deposition of amyloid-ß plaques and worsens associated cognitive deficits. This indicates that triplication of chromosome 21 genes other than APP is likely to have an important role to play in Alzheimer's disease pathogenesis in individuals who have Down syndrome. We go on to show that the effect of trisomy of chromosome 21 on amyloid-ß aggregation correlates with an unexpected shift in soluble amyloid-ß 40/42 ratio. This alteration in amyloid-ß isoform ratio occurs independently of a change in the carboxypeptidase activity of the γ-secretase complex, which cleaves the peptide from APP, or the rate of extracellular clearance of amyloid-ß. These new mechanistic insights into the role of triplication of genes on chromosome 21, other than APP, in the development of Alzheimer's disease in individuals who have Down syndrome may have implications for the treatment of this common cause of neurodegeneration.


Subject(s)
Down Syndrome/genetics , Down Syndrome/pathology , Plaque, Amyloid/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/physiology , Animals , Brain/pathology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Neurofibrillary Tangles/pathology , Plaque, Amyloid/pathology , Trisomy
2.
J Exp Med ; 215(4): 1047-1058, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29483128

ABSTRACT

One of the hallmarks of Alzheimer's disease is the presence of extracellular diffuse and fibrillar plaques predominantly consisting of the amyloid-ß (Aß) peptide. Apolipoprotein E (ApoE) influences the deposition of amyloid pathology through affecting the clearance and aggregation of monomeric Aß in the brain. In addition to influencing Aß metabolism, increasing evidence suggests that apoE influences microglial function in neurodegenerative diseases. Here, we characterize the impact that apoE has on amyloid pathology and the innate immune response in APPPS1ΔE9 and APPPS1-21 transgenic mice. We report that Apoe deficiency reduced fibrillar plaque deposition, consistent with previous studies. However, fibrillar plaques in Apoe-deficient mice exhibited a striking reduction in plaque compaction. Hyperspectral fluorescent imaging using luminescent conjugated oligothiophenes identified distinct Aß morphotypes in Apoe-deficient mice. We also observed a significant reduction in fibrillar plaque-associated microgliosis and activated microglial gene expression in Apoe-deficient mice, along with significant increases in dystrophic neurites around fibrillar plaques. Our results suggest that apoE is critical in stimulating the innate immune response to amyloid pathology.


Subject(s)
Amyloid/metabolism , Apolipoproteins E/metabolism , Microglia/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid/immunology , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/immunology , Amyloid beta-Protein Precursor/metabolism , Animals , Apolipoproteins E/immunology , Brain/immunology , Brain/metabolism , Brain/pathology , Disease Models, Animal , Humans , Immunity, Innate/immunology , Mice , Mice, Transgenic , Microglia/immunology , Microglia/pathology , Plaque, Amyloid/immunology
3.
Neuron ; 96(5): 1013-1023.e4, 2017 Dec 06.
Article in English | MEDLINE | ID: mdl-29216448

ABSTRACT

The apolipoprotein E (APOE) gene is the strongest genetic risk factor for late-onset Alzheimer disease. Previous studies suggest that reduction of apoE levels through genetic manipulation can reduce Aß pathology. However, it is not clear how reduction of apoE levels after birth would affect amyloid deposition. We utilize an antisense oligonucleotide (ASO) to reduce apoE expression in the brains of APP/PS1-21 mice homozygous for the APOE-ε4 or APOE-ε3 allele. ASO treatment starting after birth led to a significant decrease in Aß pathology when assessed at 4 months. Interestingly, ASO treatment starting at the onset of amyloid deposition led to an increase in Aß plaque size and a reduction in plaque-associated neuritic dystrophy with no change in overall plaque load. These results suggest that lowering apoE levels prior to plaque deposition can strongly affect the initiation of Aß pathology while lowering apoE after Aß seeding modulates plaque size and toxicity.


Subject(s)
Amyloid beta-Peptides , Amyloidosis/drug therapy , Apolipoproteins E/antagonists & inhibitors , Oligonucleotides, Antisense/therapeutic use , Aging/physiology , Alleles , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/biosynthesis , Amyloid beta-Protein Precursor/genetics , Amyloidosis/pathology , Animals , Apolipoprotein E3/genetics , Apolipoprotein E4/genetics , Humans , Male , Mice , Mice, Transgenic , Plaque, Amyloid/pathology , Plaque, Amyloid/prevention & control
4.
Proc Natl Acad Sci U S A ; 114(43): 11524-11529, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29073081

ABSTRACT

Variants in the gene encoding the triggering receptor expressed on myeloid cells 2 (TREM2) were recently found to increase the risk for developing Alzheimer's disease (AD). In the brain, TREM2 is predominately expressed on microglia, and its association with AD adds to increasing evidence implicating a role for the innate immune system in AD initiation and progression. Thus far, studies have found TREM2 is protective in the response to amyloid pathology while variants leading to a loss of TREM2 function impair microglial signaling and are deleterious. However, the potential role of TREM2 in the context of tau pathology has not yet been characterized. In this study, we crossed Trem2+/+ (T2+/+) and Trem2-/- (T2-/-) mice to the PS19 human tau transgenic line (PS) to investigate whether loss of TREM2 function affected tau pathology, the microglial response to tau pathology, or neurodegeneration. Strikingly, by 9 mo of age, T2-/-PS mice exhibited significantly less brain atrophy as quantified by ventricular enlargement and preserved cortical volume in the entorhinal and piriform regions compared with T2+/+PS mice. However, no TREM2-dependent differences were observed for phosphorylated tau staining or insoluble tau levels. Rather, T2-/-PS mice exhibited significantly reduced microgliosis in the hippocampus and piriform cortex compared with T2+/+PS mice. Gene expression analyses and immunostaining revealed microglial activation was significantly attenuated in T2-/-PS mice, and there were lower levels of inflammatory cytokines and astrogliosis. These unexpected findings suggest that impairing microglial TREM2 signaling reduces neuroinflammation and is protective against neurodegeneration in the setting of pure tauopathy.


Subject(s)
Inflammation/genetics , Membrane Glycoproteins/metabolism , Neurodegenerative Diseases/genetics , Receptors, Immunologic/metabolism , Tauopathies , Animals , Gene Expression Regulation/physiology , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Immunologic/genetics
6.
Sci Transl Med ; 9(386)2017 04 19.
Article in English | MEDLINE | ID: mdl-28424326

ABSTRACT

Tauopathies are a group of disorders in which the cytosolic protein tau aggregates and accumulates in cells within the brain, resulting in neurodegeneration. A promising treatment being explored for tauopathies is passive immunization with anti-tau antibodies. We previously found that administration of an anti-tau antibody to human tau transgenic mice increased the concentration of plasma tau. We further explored the effects of administering an anti-tau antibody on plasma tau. After peripheral administration of an anti-tau antibody to human patients with tauopathy and to mice expressing human tau in the central nervous system, there was a dose-dependent increase in plasma tau. In mouse plasma, we found that tau had a short half-life of 8 min that increased to more than 3 hours after administration of anti-tau antibody. As tau transgenic mice accumulated insoluble tau in the brain, brain soluble and interstitial fluid tau decreased. Administration of anti-tau antibody to tau transgenic mice that had decreased brain soluble tau and interstitial fluid tau resulted in an increase in plasma tau, but this increase was less than that observed in tau transgenic mice without these brain changes. Tau transgenic mice subjected to acute neuronal injury using 3-nitropropionic acid showed increased interstitial fluid tau and plasma tau. These data suggest that peripheral administration of an anti-tau antibody results in increased plasma tau, which correlates with the concentration of extracellular and soluble tau in the brain.


Subject(s)
Antibodies/pharmacology , Tauopathies/blood , Tauopathies/metabolism , tau Proteins/blood , tau Proteins/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Mice , Mice, Transgenic , Nitro Compounds/toxicity , Propionates/toxicity
7.
J Exp Med ; 214(5): 1227-1238, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28416651

ABSTRACT

Tauopathies are characterized by the progressive accumulation of hyperphosphorylated, aggregated forms of tau. Our laboratory has previously demonstrated that passive immunization with an anti-tau antibody, HJ8.5, decreased accumulation of pathological tau in a human P301S tau-expressing transgenic (P301S-tg) mouse model of frontotemporal dementia/tauopathy. To investigate whether the Fc domain of HJ8.5 is required for the therapeutic effect, we engineered single-chain variable fragments (scFvs) derived from HJ8.5 with variable linker lengths, all specific to human tau. Based on different binding properties, we selected two anti-tau scFvs and tested their efficacy in vivo by adeno-associated virus-mediated gene transfer to the brain of P301S-tg mice. The scFvs significantly reduced levels of hyperphosphorylated, aggregated tau in brain tissue of P301S-tg mice, associated with a decrease in detergent-soluble tau species. Interestingly, these mice showed substantial levels of scFvs in the cerebrospinal fluid without significant effects on total extracellular tau levels. Therefore, our study provides a novel strategy for anti-tau immunotherapeutics that potentially limits a detrimental proinflammatory response.


Subject(s)
Single-Chain Antibodies/immunology , Tauopathies/immunology , tau Proteins/immunology , Animals , Brain/metabolism , Dependovirus/genetics , Disease Models, Animal , Female , Gene Transfer Techniques , Hippocampus/metabolism , Male , Mice , Mice, Transgenic , Single-Chain Antibodies/genetics , Single-Chain Antibodies/physiology , Tauopathies/metabolism
8.
Mol Neurodegener ; 10: 55, 2015 Oct 26.
Article in English | MEDLINE | ID: mdl-26502977

ABSTRACT

BACKGROUND: Intracellular accumulation of tau as neurofibrillary tangles (NFTs) is the hallmark of Alzheimer's disease (AD) as well as in other tauopathies. Tau is present not only in the cytoplasm but also in the extracellular space such as cerebrospinal fluid (CSF) and brain interstitial fluid (ISF). Although clearance is one critical parameter leading to such intracellular/extracellular tau accumulation, in vivo turnover of tau has not been well characterized. The current study has attempted to precisely determine in vivo turnover rates of tau utilizing tet-off regulatable mice. In particular, we assessed intracellular tau and extracellular tau, soluble tau, insoluble tau and phosphorylated tau at certain sites utilizing a combination of in vivo microdialysis, biochemical analysis and specific ELISAs recognizing each species. To examine the effect of a tauopathy-associated mutation on tau clearance, half-lives of various tau species were compared between the mice with a FTDP-17 mutation that induces ß-sheet formation, ΔK280 mutation (pro-aggregant mice) and control mice with additional ß-sheet breaking mutations (anti-aggregant mice). RESULTS: Here we report that tau is metabolized at much slower turnover rates in vivo than in cell culture. We found that insoluble tau in pro-aggregant mice had a significantly slower half-life (t1/2 = ~34.2 days) than soluble tau (t1/2 = ~9.7 days). In contrast, soluble tau phosphorylated in the proline rich region was cleared faster than total soluble tau. When comparing pro-aggregant mice to anti-agregant mice, turnover rates of soluble tau species were not significantly different. CONCLUSIONS: The current study provides a comprehensive description of in vivo turnover of various tau species present in mice that express human tau. The turnover rate of soluble tau was not significantly altered between pro-aggregant mice and anti-aggregant mice. This suggests that altered conformation by ΔK280 does not have a major impact on clearance pathways for soluble tau. In contrast, different tau species displayed different half-lives. Turnover was significantly delayed for insoluble tau whereas it was accelerated for soluble tau phosphorylated in the proline rich region. These differences in susceptibilities to clearance suggest that aggregation and phosphorylation influences tau clearance which may be important in tau pathogenesis.


Subject(s)
Brain/metabolism , Frontotemporal Dementia/metabolism , Tauopathies/metabolism , tau Proteins/metabolism , Animals , Disease Models, Animal , Memory/physiology , Mice , Mice, Transgenic , Mutation/genetics , Tauopathies/genetics
9.
J Exp Med ; 211(13): 2487-96, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25422493

ABSTRACT

Age-related aggregation of amyloid-ß (Aß) is an upstream pathological event in Alzheimer's disease (AD) pathogenesis, and it disrupts the sleep-wake cycle. The amount of sleep declines with aging and to a greater extent in AD. Poor sleep quality and insufficient amounts of sleep have been noted in humans with preclinical evidence of AD. However, how the amount and quality of sleep affects Aß aggregation is not yet well understood. Orexins (hypocretins) initiate and maintain wakefulness, and loss of orexin-producing neurons causes narcolepsy. We tried to determine whether orexin release or secondary changes in sleep via orexin modulation affect Aß pathology. Amyloid precursor protein (APP)/Presenilin 1 (PS1) transgenic mice, in which the orexin gene is knocked out, showed a marked decrease in the amount of Aß pathology in the brain with an increase in sleep time. Focal overexpression of orexin in the hippocampus in APP/PS1 mice did not alter the total amount of sleep/wakefulness and the amount of Aß pathology. In contrast, sleep deprivation or increasing wakefulness by rescue of orexinergic neurons in APP/PS1 mice lacking orexin increased the amount of Aß pathology in the brain. Collectively, modulation of orexin and its effects on sleep appear to modulate Aß pathology in the brain.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/physiopathology , Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Sleep/physiology , Alzheimer Disease/complications , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Chronic Disease , Circadian Rhythm/physiology , Genetic Vectors/metabolism , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Mice, Knockout , Neuropeptides/deficiency , Orexins , Presenilin-1/metabolism , Promoter Regions, Genetic/genetics , Sleep Deprivation/complications , Sleep Deprivation/pathology , Sleep Deprivation/physiopathology , Wakefulness/physiology
10.
Mol Neurodegener ; 9: 20, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24893973

ABSTRACT

BACKGROUND: Recent genome-wide association studies linked variants in TREM2 to a strong increase in the odds of developing Alzheimer's disease. The mechanism by which TREM2 influences the susceptibility to Alzheimer's disease is currently unknown. TREM2 is expressed by microglia and is thought to regulate phagocytic and inflammatory microglial responses to brain pathology. Given that a single allele of variant TREM2, likely resulting in a loss of function, conferred an increased risk of developing Alzheimer's disease, we tested whether loss of one functional trem2 allele would affect Aß plaque deposition or the microglial response to Aß pathology in APPPS1-21 mice. RESULTS: There was no significant difference in Aß deposition in 3-month old or 7-month old APPPS1-21 mice expressing one or two copies of trem2. However, 3-month old mice with one copy of trem2 exhibited a marked decrease in the number and size of plaque-associated microglia. While there were no statistically significant differences in cytokine levels or markers of microglial activation in 3- or 7-month old animals, there were trends towards decreased expression of NOS2, C1qa, and IL1a in 3-month old TREM2+/- vs. TREM2+/+ mice. CONCLUSIONS: Loss of a single copy of trem2 had no effect on Aß pathology, but altered the morphological phenotype of plaque-associated microglia. These data suggest that TREM2 is important for the microglial response to Aß deposition but that a 50% decrease inTREM2 expression does not affect Aß plaque burden.


Subject(s)
Alzheimer Disease , Membrane Glycoproteins/genetics , Microglia/metabolism , Plaque, Amyloid/genetics , Receptors, Immunologic/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Female , Heterozygote , Male , Mice , Mice, Transgenic , Microglia/pathology , Plaque, Amyloid/pathology , Presenilin-1/genetics , Real-Time Polymerase Chain Reaction
11.
J Exp Med ; 211(3): 387-93, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24534188

ABSTRACT

Tau is primarily a cytoplasmic protein that stabilizes microtubules. However, it is also found in the extracellular space of the brain at appreciable concentrations. Although its presence there may be relevant to the intercellular spread of tau pathology, the cellular mechanisms regulating tau release into the extracellular space are not well understood. To test this in the context of neuronal networks in vivo, we used in vivo microdialysis. Increasing neuronal activity rapidly increased the steady-state levels of extracellular tau in vivo. Importantly, presynaptic glutamate release is sufficient to drive tau release. Although tau release occurred within hours in response to neuronal activity, the elimination rate of tau from the extracellular compartment and the brain is slow (half-life of ∼11 d). The in vivo results provide one mechanism underlying neuronal tau release and may link trans-synaptic spread of tau pathology with synaptic activity itself.


Subject(s)
Brain/metabolism , Extracellular Space/metabolism , Neurons/metabolism , Synaptic Transmission/physiology , Tauopathies/physiopathology , tau Proteins/metabolism , Analysis of Variance , Animals , Electroencephalography , Enzyme-Linked Immunosorbent Assay , Female , Glutamic Acid/metabolism , Half-Life , Kinetics , Luciferases , Male , Mice , Mice, Transgenic , Microdialysis , Tetrodotoxin
12.
J Neurosci ; 33(31): 12887-97, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23904623

ABSTRACT

Tau, a microtubule-associated protein, is implicated in the pathogenesis of Alzheimer's Disease (AD) in regard to both neurofibrillary tangle formation and neuronal network hyperexcitability. The genetic ablation of tau substantially reduces hyperexcitability in AD mouse lines, induced seizure models, and genetic in vivo models of epilepsy. These data demonstrate that tau is an important regulator of network excitability. However, developmental compensation in the genetic tau knock-out line may account for the protective effect against seizures. To test the efficacy of a tau reducing therapy for disorders with a detrimental hyperexcitability profile in adult animals, we identified antisense oligonucleotides that selectively decrease endogenous tau expression throughout the entire mouse CNS--brain and spinal cord tissue, interstitial fluid, and CSF--while having no effect on baseline motor or cognitive behavior. In two chemically induced seizure models, mice with reduced tau protein had less severe seizures than control mice. Total tau protein levels and seizure severity were highly correlated, such that those mice with the most severe seizures also had the highest levels of tau. Our results demonstrate that endogenous tau is integral for regulating neuronal hyperexcitability in adult animals and suggest that an antisense oligonucleotide reduction of tau could benefit those with epilepsy and perhaps other disorders associated with tau-mediated neuronal hyperexcitability.


Subject(s)
Anticonvulsants/therapeutic use , Oligonucleotides, Antisense/therapeutic use , Seizures/prevention & control , tau Proteins/genetics , Age Factors , Animals , Central Nervous System/drug effects , Central Nervous System/metabolism , Convulsants/toxicity , Disease Models, Animal , Gait Disorders, Neurologic/drug therapy , Gait Disorders, Neurologic/etiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Infusions, Intraventricular , Lactic Acid/metabolism , Locomotion/genetics , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microdialysis , Pentylenetetrazole/toxicity , Picrotoxin/toxicity , Seizures/chemically induced , Seizures/genetics , tau Proteins/chemistry , tau Proteins/metabolism
13.
J Exp Med ; 209(12): 2149-56, 2012 Nov 19.
Article in English | MEDLINE | ID: mdl-23129750

ABSTRACT

The apolipoprotein E (APOE) ε4 allele is the strongest genetic risk factor for Alzheimer's disease (AD). The influence of apoE on amyloid ß (Aß) accumulation may be the major mechanism by which apoE affects AD. ApoE interacts with Aß and facilitates Aß fibrillogenesis in vitro. In addition, apoE is one of the protein components in plaques. We hypothesized that certain anti-apoE antibodies, similar to certain anti-Aß antibodies, may have antiamyloidogenic effects by binding to apoE in the plaques and activating microglia-mediated amyloid clearance. To test this hypothesis, we developed several monoclonal anti-apoE antibodies. Among them, we administered HJ6.3 antibody intraperitoneally to 4-mo-old male APPswe/PS1ΔE9 mice weekly for 14 wk. HJ6.3 dramatically decreased amyloid deposition by 60-80% and significantly reduced insoluble Aß40 and Aß42 levels. Short-term treatment with HJ6.3 resulted in strong changes in microglial responses around Aß plaques. Collectively, these results suggest that anti-apoE immunization may represent a novel AD therapeutic strategy and that other proteins involved in Aß binding and aggregation might also be a target for immunotherapy. Our data also have important broader implications for other amyloidosis. Immunotherapy to proteins tightly associated with misfolded proteins might open up a new treatment option for many protein misfolding diseases.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloidosis/prevention & control , Antibodies, Monoclonal/pharmacology , Apolipoproteins E/immunology , Brain Diseases/prevention & control , Immunotherapy/methods , Amyloidosis/immunology , Amyloidosis/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/blood , Apolipoproteins E/metabolism , Brain Diseases/immunology , Brain Diseases/metabolism , Enzyme-Linked Immunosorbent Assay , Immunoblotting , Immunoprecipitation , Male , Mice , Mice, Transgenic , Statistics, Nonparametric
14.
Int J Clin Exp Pathol ; 5(6): 482-95, 2012.
Article in English | MEDLINE | ID: mdl-22949930

ABSTRACT

Alzheimer's disease is characterized in part by extracellular aggregation of the amyloid-ß peptide in the form of diffuse and fibrillar plaques in the brain. Electron microscopy (EM) has made an important contribution in understanding of the structure of amyloid plaques in humans. Classical EM studies have revealed the architecture of the fibrillar core, characterized the progression of neuritic changes, and have identified the neurofibrillary tangles formed by paired helical filaments (PHF) in degenerating neurons. Clinical data has strongly correlated cognitive impairment in AD with the substantial synapse loss observed in these early ultrastructural studies. Animal models of AD-type brain amyloidosis have provided excellent opportunities to study amyloid and neuritic pathology in detail and establish the role of neurons and glia in plaque formation. Transgenic mice overexpressing mutant amyloid precursor protein (APP) alone with or without mutant presenilin 1 (PS1), have shown that brain amyloid plaque development and structure grossly recapitulate classical findings in humans. Transgenic APP/PS1 mice expressing human apolioprotein E isoforms also develop amyloid plaque deposition. However no ultrastructural data has been reported for these animals. Here we show results from detailed EM analysis of amyloid plaques in APP/PS1 mice expressing human isoforms of ApoE and compare these findings with EM data in other transgenic models and in human AD. Our results show that similar to other transgenic animals, APP/PS1 mice expressing human ApoE isoforms share all major cellular and subcellular degenerative features and highlight the identity of the cellular elements involved in Aß deposition and neuronal degeneration.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/metabolism , Apolipoproteins E/metabolism , Plaque, Amyloid/ultrastructure , Presenilin-1/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/ultrastructure , Amyloidosis/pathology , Animals , Apolipoproteins E/ultrastructure , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Mutation , Neurons/metabolism , Neurons/ultrastructure , Plaque, Amyloid/metabolism , Presenilin-1/ultrastructure , Protein Isoforms
15.
Sci Transl Med ; 4(150): 150ra122, 2012 Sep 05.
Article in English | MEDLINE | ID: mdl-22956200

ABSTRACT

Aggregation of ß-amyloid (Aß) in the brain begins to occur years before the clinical onset of Alzheimer's disease (AD). Before Aß aggregation, concentrations of extracellular soluble Aß in the interstitial fluid (ISF) space of the brain, which are regulated by neuronal activity and the sleep-wake cycle, correlate with the amount of Aß deposition in the brain seen later. The amount and quality of sleep decline with normal aging and to a greater extent in AD patients. How sleep quality as well as the diurnal fluctuation in Aß change with age and Aß aggregation is not well understood. We report a normal sleep-wake cycle and diurnal fluctuation in ISF Aß in the brain of the APPswe/PS1δE9 mouse model of AD before Aß plaque formation. After plaque formation, the sleep-wake cycle markedly deteriorated and diurnal fluctuation of ISF Aß dissipated. As in mice, diurnal fluctuation of cerebrospinal fluid Aß in young adult humans with presenilin mutations was also markedly attenuated after Aß plaque formation. Virtual elimination of Aß deposits in the mouse brain by active immunization with Aß(42) normalized the sleep-wake cycle and the diurnal fluctuation of ISF Aß. These data suggest that Aß aggregation disrupts the sleep-wake cycle and diurnal fluctuation of Aß. Sleep-wake behavior and diurnal fluctuation of Aß in the central nervous system may be functional and biochemical indicators, respectively, of Aß-associated pathology.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Circadian Rhythm/physiology , Sleep/physiology , Wakefulness/physiology , Alzheimer Disease/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Animals , Disease Models, Animal , Extracellular Fluid/metabolism , Genes, Dominant/genetics , Hippocampus/metabolism , Hippocampus/pathology , Humans , Immunization , Lactates/metabolism , Mice , Mice, Transgenic , Mutation/genetics , Neostriatum/metabolism , Neostriatum/pathology , Presenilins/genetics , Time Factors
16.
Proc Natl Acad Sci U S A ; 109(38): 15502-7, 2012 Sep 18.
Article in English | MEDLINE | ID: mdl-22927427

ABSTRACT

The apolipoprotein E (APOE)-ε4 allele is the strongest genetic risk factor for late-onset, sporadic Alzheimer's disease, likely increasing risk by altering amyloid-ß (Aß) accumulation. We recently demonstrated that the low-density lipoprotein receptor (LDLR) is a major apoE receptor in the brain that strongly regulates amyloid plaque deposition. In the current study, we sought to understand the mechanism by which LDLR regulates Aß accumulation by altering Aß clearance from brain interstitial fluid. We hypothesized that increasing LDLR levels enhances blood-brain barrier-mediated Aß clearance, thus leading to reduced Aß accumulation. Using the brain Aß efflux index method, we found that blood-brain barrier-mediated clearance of exogenously administered Aß is enhanced with LDLR overexpression. We next developed a method to directly assess the elimination of centrally derived, endogenous Aß into the plasma of mice using an anti-Aß antibody that prevents degradation of plasma Aß, allowing its rate of appearance from the brain to be measured. Using this plasma Aß accumulation technique, we found that LDLR overexpression enhances brain-to-blood Aß transport. Together, our results suggest a unique mechanism by which LDLR regulates brain-to-blood Aß clearance, which may serve as a useful therapeutic avenue in targeting Aß clearance from the brain.


Subject(s)
Amyloidosis/metabolism , Apolipoprotein E4/genetics , Receptors, LDL/biosynthesis , Alleles , Amyloid beta-Peptides/metabolism , Animals , Blood-Brain Barrier , Brain/metabolism , Disease Models, Animal , Insulin/metabolism , Kinetics , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Microdialysis , Transgenes
17.
J Neurosci ; 32(13): 4334-40, 2012 Mar 28.
Article in English | MEDLINE | ID: mdl-22457485

ABSTRACT

Brain region-specific deposition of extracellular amyloid plaques principally composed of aggregated amyloid-ß (Aß) peptide is a pathological signature of Alzheimer's disease (AD). Recent human neuroimaging data suggest that resting-state functional connectivity strength is reduced in patients with AD, cognitively normal elderly harboring elevated amyloid burden, and in advanced aging. Interestingly, there exists a striking spatial correlation between functional connectivity strength in cognitively normal adults and the location of Aß plaque deposition in AD. However, technical limitations have heretofore precluded examination of the relationship between functional connectivity, Aß deposition, and normal aging in mouse models. Using a novel functional connectivity optical intrinsic signal (fcOIS) imaging technique, we demonstrate that Aß deposition is associated with significantly reduced bilateral functional connectivity in multiple brain regions of older APP/PS1 transgenic mice. The amount of Aß deposition in each brain region was associated with the degree of local, age-related bilateral functional connectivity decline. Normal aging was associated with reduced bilateral functional connectivity specifically in retrosplenial cortex. Furthermore, we found that the magnitude of regional bilateral functional correlation in young APP/PS1 mice before Aß plaque formation was proportional to the amount of region-specific plaque deposition seen later in older APP/PS1 mice. Together, these findings suggest that Aß deposition and normal aging are associated with region-specific disruption of functional connectivity and that the magnitude of local bilateral functional connectivity predicts regional vulnerability to subsequent Aß deposition in mouse brain.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/physiopathology , Functional Neuroimaging/statistics & numerical data , Neural Pathways/physiopathology , Plaque, Amyloid/metabolism , Aging/metabolism , Aging/physiology , Amyloidosis/metabolism , Amyloidosis/physiopathology , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Functional Neuroimaging/methods , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , Neural Pathways/metabolism
18.
J Neurosci ; 31(49): 18007-12, 2011 Dec 07.
Article in English | MEDLINE | ID: mdl-22159114

ABSTRACT

The ε4 allele of the apolipoprotein E (APOE) gene is the strongest genetic risk factor for Alzheimer's disease (AD). Evidence suggests that the effect of apoE isoforms on amyloid-ß (Aß) accumulation in the brain plays a critical role in AD pathogenesis. Like in humans, apoE4 expression in animal models that develop Aß amyloidosis results in greater Aß and amyloid deposition than with apoE3 expression. However, whether decreasing levels of apoE3 or apoE4 would promote or attenuate Aß-related pathology has not been directly addressed. To determine the effect of decreasing human apoE levels on Aß accumulation in vivo, we generated human APOE isoform haploinsufficient mouse models by crossing APPPS1-21 mice with APOE isoform knock-in mice. By genetically manipulating APOE gene dosage, we demonstrate that decreasing human apoE levels, regardless of isoform status, results in significantly decreased amyloid plaque deposition and microglial activation. These differences in amyloid load between apoE3- and apoE4-expressing mice were not due to apoE4 protein being present at lower levels than apoE3. These data suggest that current therapeutic strategies to increase apoE levels without altering its lipidation state may actually worsen Aß amyloidosis, while increasing apoE degradation or inhibiting its synthesis may be a more effective treatment approach.


Subject(s)
Amyloid/metabolism , Amyloidosis/genetics , Amyloidosis/metabolism , Apolipoproteins E/deficiency , Haploinsufficiency/genetics , Amyloid beta-Protein Precursor/genetics , Amyloidosis/pathology , Animals , Apolipoproteins E/genetics , Brain/metabolism , Brain/pathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/genetics , Humans , Mice , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/metabolism , Plaque, Amyloid/pathology , Presenilin-1/genetics , Protein Isoforms/genetics
19.
Proc Natl Acad Sci U S A ; 108(47): 19054-9, 2011 Nov 22.
Article in English | MEDLINE | ID: mdl-22058226

ABSTRACT

Hypoxic-ischemic (H-I) injury to the developing brain is a significant cause of morbidity and mortality in humans. Other than hypothermia, there is no effective treatment to prevent or lessen the consequences of neonatal H-I. Increased expression of the NAD synthesizing enzyme nicotinamide mononucleotide adenylyl transferase 1 (Nmnat1) has been shown to be neuroprotective against axonal injury in the peripheral nervous system. To investigate the neuroprotective role of Nmnat1 against acute neurodegeneration in the developing CNS, we exposed wild-type mice and mice overexpressing Nmnat1 in the cytoplasm (cytNmnat1-Tg mice) to a well-characterized model of neonatal H-I brain injury. As early as 6 h after H-I, cytNmnat1-Tg mice had strikingly less injury detected by MRI. CytNmnat1-Tg mice had markedly less injury in hippocampus, cortex, and striatum than wild-type mice as assessed by loss of tissue volume 7 d days after H-I. The dramatic protection mediated by cytNmnat1 is not mediated through modulating caspase3-dependent cell death in cytNmnat1-Tg brains. CytNmnat1 protected neuronal cell bodies and processes against NMDA-induced excitotoxicity, whereas caspase inhibition or B-cell lymphoma-extra large (Bcl-XL) protein overexpression had no protective effects in cultured cortical neurons. These results suggest that cytNmnat1 protects against neonatal HI-induced CNS injury by inhibiting excitotoxicity-induced, caspase-independent injury to neuronal processes and cell bodies. As such, the Nmnat1 protective pathway could be a useful therapeutic target for acute and chronic neurodegenerative insults mediated by excitotoxicity.


Subject(s)
Cell Death/physiology , Hypoxia-Ischemia, Brain/complications , Necrosis/metabolism , Nerve Degeneration/enzymology , Nerve Degeneration/etiology , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Analysis of Variance , Animals , Animals, Newborn , Chromatography, High Pressure Liquid , Humans , Hypoxia-Ischemia, Brain/pathology , Immunohistochemistry , L-Lactate Dehydrogenase/metabolism , Magnetic Resonance Imaging , Mice , Nerve Degeneration/pathology
20.
J Neurosci ; 31(37): 13110-7, 2011 Sep 14.
Article in English | MEDLINE | ID: mdl-21917794

ABSTRACT

Although tau is a cytoplasmic protein, it is also found in brain extracellular fluids, e.g., CSF. Recent findings suggest that aggregated tau can be transferred between cells and extracellular tau aggregates might mediate spread of tau pathology. Despite these data, details of whether tau is normally released into the brain interstitial fluid (ISF), its concentration in ISF in relation to CSF, and whether ISF tau is influenced by its aggregation are unknown. To address these issues, we developed a microdialysis technique to analyze monomeric ISF tau levels within the hippocampus of awake, freely moving mice. We detected tau in ISF of wild-type mice, suggesting that tau is released in the absence of neurodegeneration. ISF tau was significantly higher than CSF tau and their concentrations were not significantly correlated. Using P301S human tau transgenic mice (P301S tg mice), we found that ISF tau is fivefold higher than endogenous murine tau, consistent with its elevated levels of expression. However, following the onset of tau aggregation, monomeric ISF tau decreased markedly. Biochemical analysis demonstrated that soluble tau in brain homogenates decreased along with the deposition of insoluble tau. Tau fibrils injected into the hippocampus decreased ISF tau, suggesting that extracellular tau is in equilibrium with extracellular or intracellular tau aggregates. This technique should facilitate further studies of tau secretion, spread of tau pathology, the effects of different disease states on ISF tau, and the efficacy of experimental treatments.


Subject(s)
Aging/metabolism , Extracellular Fluid/metabolism , Hippocampus/metabolism , Microdialysis/methods , tau Proteins/genetics , tau Proteins/metabolism , Aging/cerebrospinal fluid , Animals , Female , Humans , Male , Mice , Mice, Transgenic , Models, Neurological , Solubility , tau Proteins/administration & dosage , tau Proteins/cerebrospinal fluid , tau Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...