Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Nature ; 615(7953): 668-677, 2023 03.
Article in English | MEDLINE | ID: mdl-36890231

ABSTRACT

Extracellular deposition of amyloid-ß as neuritic plaques and intracellular accumulation of hyperphosphorylated, aggregated tau as neurofibrillary tangles are two of the characteristic hallmarks of Alzheimer's disease1,2. The regional progression of brain atrophy in Alzheimer's disease highly correlates with tau accumulation but not amyloid deposition3-5, and the mechanisms of tau-mediated neurodegeneration remain elusive. Innate immune responses represent a common pathway for the initiation and progression of some neurodegenerative diseases. So far, little is known about the extent or role of the adaptive immune response and its interaction with the innate immune response in the presence of amyloid-ß or tau pathology6. Here we systematically compared the immunological milieux in the brain of mice with amyloid deposition or tau aggregation and neurodegeneration. We found that mice with tauopathy but not those with amyloid deposition developed a unique innate and adaptive immune response and that depletion of microglia or T cells blocked tau-mediated neurodegeneration. Numbers of T cells, especially those of cytotoxic T cells, were markedly increased in areas with tau pathology in mice with tauopathy and in the Alzheimer's disease brain. T cell numbers correlated with the extent of neuronal loss, and the cells dynamically transformed their cellular characteristics from activated to exhausted states along with unique TCR clonal expansion. Inhibition of interferon-γ and PDCD1 signalling both significantly ameliorated brain atrophy. Our results thus reveal a tauopathy- and neurodegeneration-related immune hub involving activated microglia and T cell responses, which could serve as therapeutic targets for preventing neurodegeneration in Alzheimer's disease and primary tauopathies.


Subject(s)
Brain , Microglia , Neurofibrillary Tangles , T-Lymphocytes , Tauopathies , Animals , Mice , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Brain/immunology , Brain/metabolism , Brain/pathology , Microglia/immunology , Microglia/metabolism , Neurofibrillary Tangles/immunology , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , tau Proteins/immunology , tau Proteins/metabolism , Tauopathies/immunology , Tauopathies/metabolism , Tauopathies/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Plaque, Amyloid/immunology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology , Clone Cells/immunology , Clone Cells/metabolism , Clone Cells/pathology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Immunity, Innate
2.
Nature ; 549(7673): 523-527, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28959956

ABSTRACT

APOE4 is the strongest genetic risk factor for late-onset Alzheimer disease. ApoE4 increases brain amyloid-ß pathology relative to other ApoE isoforms. However, whether APOE independently influences tau pathology, the other major proteinopathy of Alzheimer disease and other tauopathies, or tau-mediated neurodegeneration, is not clear. By generating P301S tau transgenic mice on either a human ApoE knock-in (KI) or ApoE knockout (KO) background, here we show that P301S/E4 mice have significantly higher tau levels in the brain and a greater extent of somatodendritic tau redistribution by three months of age compared with P301S/E2, P301S/E3, and P301S/EKO mice. By nine months of age, P301S mice with different ApoE genotypes display distinct phosphorylated tau protein (p-tau) staining patterns. P301S/E4 mice develop markedly more brain atrophy and neuroinflammation than P301S/E2 and P301S/E3 mice, whereas P301S/EKO mice are largely protected from these changes. In vitro, E4-expressing microglia exhibit higher innate immune reactivity after lipopolysaccharide treatment. Co-culturing P301S tau-expressing neurons with E4-expressing mixed glia results in a significantly higher level of tumour-necrosis factor-α (TNF-α) secretion and markedly reduced neuronal viability compared with neuron/E2 and neuron/E3 co-cultures. Neurons co-cultured with EKO glia showed the greatest viability with the lowest level of secreted TNF-α. Treatment of P301S neurons with recombinant ApoE (E2, E3, E4) also leads to some neuronal damage and death compared with the absence of ApoE, with ApoE4 exacerbating the effect. In individuals with a sporadic primary tauopathy, the presence of an ε4 allele is associated with more severe regional neurodegeneration. In individuals who are positive for amyloid-ß pathology with symptomatic Alzheimer disease who usually have tau pathology, ε4-carriers demonstrate greater rates of disease progression. Our results demonstrate that ApoE affects tau pathogenesis, neuroinflammation, and tau-mediated neurodegeneration independently of amyloid-ß pathology. ApoE4 exerts a 'toxic' gain of function whereas the absence of ApoE is protective.


Subject(s)
Apolipoprotein E4/metabolism , Apolipoprotein E4/toxicity , Tauopathies/metabolism , Tauopathies/pathology , tau Proteins/metabolism , Alleles , Animals , Apolipoprotein E4/deficiency , Apolipoprotein E4/genetics , Cell Survival/drug effects , Coculture Techniques , Disease Models, Animal , Disease Progression , Gene Knock-In Techniques , Genotype , Humans , Immunity, Innate , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Knockout , Mice, Transgenic , Microglia/immunology , Microglia/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Phosphoproteins/analysis , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Tauopathies/genetics , Tumor Necrosis Factor-alpha/metabolism , tau Proteins/genetics
3.
Ann Neurol ; 85(2): 291-295, 2019 02.
Article in English | MEDLINE | ID: mdl-30597615

ABSTRACT

Obstructive sleep apnea (OSA) increases risk of dementia, a relationship that may be mediated by amyloid-ß (Aß) and downstream Alzheimer disease pathology. We previously showed that OSA may impair Aß clearance and affect the relationship between slow wave activity (SWA) and Aß. In this study, SWA and CSF Aß were measured in participants with OSA before and 1 to 4 months after treatment. OSA treatment increased SWA, and SWA was significantly correlated with lower Aß after treatment. Greater improvement in OSA was associated with greater decreases in Aß. We propose a model whereby OSA treatment may affect both Aß release and clearance. Ann Neurol 2018 ANN NEUROL 2019;85:291-295.


Subject(s)
Continuous Positive Airway Pressure , Sleep Apnea, Obstructive/therapy , Sleep, Slow-Wave , Aged , Amyloid beta-Peptides/cerebrospinal fluid , Female , Humans , Male , Middle Aged , Peptide Fragments/cerebrospinal fluid , Sleep Apnea, Obstructive/cerebrospinal fluid , Sleep Deprivation/cerebrospinal fluid , Treatment Outcome , tau Proteins/cerebrospinal fluid
4.
Ann Neurol ; 80(1): 154-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27129429

ABSTRACT

We hypothesized that one mechanism underlying the association between obstructive sleep apnea (OSA) and Alzheimer's disease is OSA leading to decreased slow wave activity (SWA), increased synaptic activity, decreased glymphatic clearance, and increased amyloid-ß. Polysomnography and lumbar puncture were performed in OSA and control groups. SWA negatively correlated with cerebrospinal fluid (CSF) amyloid-ß-40 among controls and was decreased in the OSA group. Unexpectedly, amyloid-ß-40 was decreased in the OSA group. Other neuronally derived proteins, but not total protein, were also decreased in the OSA group, suggesting that OSA may affect the interaction between interstitial and cerebrospinal fluid. Ann Neurol 2016;80:154-159.


Subject(s)
Amyloid beta-Peptides/cerebrospinal fluid , Central Nervous System/metabolism , Nerve Tissue Proteins/cerebrospinal fluid , Peptide Fragments/cerebrospinal fluid , Sleep Apnea, Obstructive/cerebrospinal fluid , Aged , Case-Control Studies , Female , Humans , Male , Middle Aged , Polysomnography
5.
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
6.
J Neurosci ; 32(43): 15181-92, 2012 Oct 24.
Article in English | MEDLINE | ID: mdl-23100439

ABSTRACT

Alzheimer's disease (AD) is the most common progressive neurodegenerative disorder causing dementia. Massive deposition of amyloid ß peptide (Aß) as senile plaques in the brain is the pathological hallmark of AD, but oligomeric, soluble forms of Aß have been implicated as the synaptotoxic component. The apolipoprotein E ε 4 (apoE ε4) allele is known to be a genetic risk factor for developing AD. However, it is still unknown how apoE impacts the process of Aß oligomerization. Here, we found that the level of Aß oligomers in APOE ε4/ε4 AD patient brains is 2.7 times higher than those in APOE ε3/ε3 AD patient brains, matched for total plaque burden, suggesting that apoE4 impacts the metabolism of Aß oligomers. To test this hypothesis, we examined the effect of apoE on Aß oligomer formation. Using both synthetic Aß and a split-luciferase method for monitoring Aß oligomers, we observed that apoE increased the level of Aß oligomers in an isoform-dependent manner (E2 < E3 < E4). This effect appears to be dependent on the ApoE C-terminal domain. Moreover, these results were confirmed using endogenous apoE isolated from the TBS-soluble fraction of human brain, which increased the formation of Aß oligomers. Together, these data show that lipidated apoE, especially apoE4, increases Aß oligomers in the brain. Higher levels of Aß oligomers in the brains of APOE ε4/ε4 carriers compared with APOE ε3/ε3 carriers may increase the loss of dendritic spines and accelerate memory impairments, leading to earlier cognitive decline in AD.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Brain/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/pharmacology , Analysis of Variance , Apolipoprotein E2/genetics , Apolipoprotein E2/metabolism , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Astrocytes/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Green Fluorescent Proteins/genetics , HEK293 Cells/drug effects , HEK293 Cells/metabolism , Humans , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Male , Morpholinos/pharmacology , Peptide Fragments/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transfection
7.
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
8.
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
9.
J Clin Invest ; 118(12): 4002-13, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19033669

ABSTRACT

Neurotoxic amyloid beta peptide (Abeta) accumulates in the brains of individuals with Alzheimer disease (AD). The APOE4 allele is a major risk factor for sporadic AD and has been associated with increased brain parenchymal and vascular amyloid burden. How apoE isoforms influence Abeta accumulation in the brain has, however, remained unclear. Here, we have shown that apoE disrupts Abeta clearance across the mouse blood-brain barrier (BBB) in an isoform-specific manner (specifically, apoE4 had a greater disruptive effect than either apoE3 or apoE2). Abeta binding to apoE4 redirected the rapid clearance of free Abeta40/42 from the LDL receptor-related protein 1 (LRP1) to the VLDL receptor (VLDLR), which internalized apoE4 and Abeta-apoE4 complexes at the BBB more slowly than LRP1. In contrast, apoE2 and apoE3 as well as Abeta-apoE2 and Abeta-apoE3 complexes were cleared at the BBB via both VLDLR and LRP1 at a substantially faster rate than Abeta-apoE4 complexes. Astrocyte-secreted lipo-apoE2, lipo-apoE3, and lipo-apoE4 as well as their complexes with Abeta were cleared at the BBB by mechanisms similar to those of their respective lipid-poor isoforms but at 2- to 3-fold slower rates. Thus, apoE isoforms differentially regulate Abeta clearance from the brain, and this might contribute to the effects of APOE genotype on the disease process in both individuals with AD and animal models of AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apolipoprotein E3/metabolism , Apolipoprotein E4/metabolism , Astrocytes/metabolism , Blood-Brain Barrier/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Animals , Apolipoprotein E3/genetics , Apolipoprotein E4/genetics , Disease Models, Animal , Humans , Low Density Lipoprotein Receptor-Related Protein-1 , Male , Mice , Protein Isoforms/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
10.
Neuron ; 48(6): 913-22, 2005 Dec 22.
Article in English | MEDLINE | ID: mdl-16364896

ABSTRACT

Aggregation of the amyloid-beta (Abeta) peptide in the extracellular space of the brain is central to Alzheimer's disease pathogenesis. Abeta aggregation is concentration dependent and brain region specific. Utilizing in vivo microdialysis concurrently with field potential recordings, we demonstrate that Abeta levels in the brain interstitial fluid are dynamically and directly influenced by synaptic activity on a timescale of minutes to hours. Using an acute brain slice model, we show that the rapid effects of synaptic activity on Abeta levels are primarily related to synaptic vesicle exocytosis. These results suggest that synaptic activity may modulate a neurodegenerative disease process, in this case by influencing Abeta metabolism and ultimately region-specific Abeta deposition. The findings also have important implications for treatment development.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/metabolism , Extracellular Fluid/metabolism , Presynaptic Terminals/metabolism , Synaptic Transmission/physiology , Action Potentials/drug effects , Action Potentials/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Exocytosis/physiology , Female , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microdialysis , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Organ Culture Techniques , Patch-Clamp Techniques , Perforant Pathway/physiology , Plaque, Amyloid/metabolism , Synaptic Transmission/drug effects , Synaptic Vesicles/metabolism
11.
J Neurosci ; 28(52): 14156-64, 2008 Dec 24.
Article in English | MEDLINE | ID: mdl-19109498

ABSTRACT

Aggregation of amyloid-beta (Abeta) peptide in the brain in the form of neuritic plaques and cerebral amyloid angiopathy (CAA) is a key feature of Alzheimer's disease (AD). Microglial cells surround aggregated Abeta and are believed to play a role in AD pathogenesis. A therapy for AD that has entered clinical trials is the administration of anti-Abeta antibodies. One mechanism by which certain anti-Abeta antibodies have been proposed to exert their effects is via antibody-mediated microglial activation. Whether, when, or to what extent microglial activation occurs after systemic administration of anti-Abeta antibodies has not been fully assessed. We administered an anti-Abeta antibody (m3D6) that binds aggregated Abeta to PDAPP mice, an AD mouse model that was bred to contain fluorescent microglia. Three days after systemic administration of m3D6, there was a marked increase in both the number of microglial cells and processes per cell visualized in vivo by multiphoton microscopy. These changes required the Fc domain of m3D6 and were not observed with an antibody specific to soluble Abeta. These findings demonstrate that some effects of antibodies that recognize aggregated Abeta are rapid, involve microglia, and provide insight into the mechanism of action of a specific passive immunotherapy for AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid/immunology , Antibodies/therapeutic use , Microglia/drug effects , Age Factors , Alkenes , Amyloid beta-Protein Precursor/genetics , Animals , Benzene Derivatives , CX3C Chemokine Receptor 1 , Calcium-Binding Proteins/metabolism , Cell Count/methods , Cell Movement/drug effects , Cell Movement/genetics , Cerebral Amyloid Angiopathy , Disease Models, Animal , Green Fluorescent Proteins/genetics , Leukocyte Common Antigens/metabolism , Mice , Mice, Transgenic , Microfilament Proteins , Microglia/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Receptors, Chemokine/genetics , Stilbenes
12.
Mol Neurodegener ; 14(1): 37, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31623648

ABSTRACT

BACKGROUND: The apolipoprotein E (APOE) gene is the strongest genetic risk factor for late-onset Alzheimer disease (AD). ApoE is produced by both astrocytes and microglia in the brain, whereas hepatocytes produce the majority of apoE found in the periphery. Studies using APOE knock-in and transgenic mice have demonstrated a strong isoform-dependent effect of apoE on the accumulation of amyloid-ß (Aß) deposition in the brain in the form of both Aß-containing amyloid plaques and cerebral amyloid angiopathy. However, the specific contributions of different apoE pools to AD pathogenesis remain unknown. METHODS: We have begun to address these questions by generating new lines of APOE knock-in (APOE-KI) mice (ε2/ε2, ε3/ε3, and ε4/ε4) where the exons in the coding region of APOE are flanked by loxP sites, allowing for cell type-specific manipulation of gene expression. We assessed these mice both alone and after crossing them with mice with amyloid deposition in the brain. Using biochemical and histological methods. We also investigated how removal of APOE expression from hepatocytes affected cerebral amyloid deposition. RESULTS: As in other APOE knock-in mice, apoE protein was present predominantly in astrocytes in the brain under basal conditions and was also detected in reactive microglia surrounding amyloid plaques. Primary cultured astrocytes and microglia from the APOE-KI mice secreted apoE in lipoprotein particles of distinct size distribution upon native gel analysis with microglial particles being substantially smaller than the HDL-like particles secreted by astrocytes. Crossing of APP/PS1 transgenic mice to the different APOE-KI mice recapitulated the previously described isoform-specific effect (ε4 > ε3) on amyloid plaque and Aß accumulation. Deletion of APOE in hepatocytes did not alter brain apoE levels but did lead to a marked decrease in plasma apoE levels and changes in plasma lipid profile. Despite these changes in peripheral apoE and on plasma lipids, cerebral accumulation of amyloid plaques in APP/PS1 mice was not affected. CONCLUSIONS: Altogether, these new knock-in strains offer a novel and dynamic tool to study the role of APOE in AD pathogenesis in a spatially and temporally controlled manner.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Apolipoproteins E/genetics , Brain/metabolism , Amyloid beta-Peptides/metabolism , Animals , Astrocytes/metabolism , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/pathology , Disease Models, Animal , Mice, Transgenic , Microglia/metabolism , Plaque, Amyloid/pathology
13.
Science ; 363(6429): 880-884, 2019 02 22.
Article in English | MEDLINE | ID: mdl-30679382

ABSTRACT

The sleep-wake cycle regulates interstitial fluid (ISF) and cerebrospinal fluid (CSF) levels of ß-amyloid (Aß) that accumulates in Alzheimer's disease (AD). Furthermore, chronic sleep deprivation (SD) increases Aß plaques. However, tau, not Aß, accumulation appears to drive AD neurodegeneration. We tested whether ISF/CSF tau and tau seeding and spreading were influenced by the sleep-wake cycle and SD. Mouse ISF tau was increased ~90% during normal wakefulness versus sleep and ~100% during SD. Human CSF tau also increased more than 50% during SD. In a tau seeding-and-spreading model, chronic SD increased tau pathology spreading. Chemogenetically driven wakefulness in mice also significantly increased both ISF Aß and tau. Thus, the sleep-wake cycle regulates ISF tau, and SD increases ISF and CSF tau as well as tau pathology spreading.


Subject(s)
Brain/metabolism , Circadian Rhythm , Extracellular Fluid/chemistry , Sleep Deprivation/metabolism , Sleep/physiology , Wakefulness/physiology , tau Proteins/analysis , tau Proteins/cerebrospinal fluid , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/metabolism , Animals , Extracellular Fluid/metabolism , Female , Male , Mice , Mice, Transgenic , Sleep Deprivation/cerebrospinal fluid , Wakefulness/genetics , tau Proteins/metabolism
14.
J Clin Invest ; 115(11): 3285-90, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16239972

ABSTRACT

Accumulation of amyloid-beta (Abeta) within extracellular spaces of the brain is a hallmark of Alzheimer disease (AD). In sporadic, late-onset AD, there is little evidence for increased Abeta production, suggesting that decreased elimination from the brain may contribute to elevated levels of Abeta and plaque formation. Efflux transport of Abeta across the blood-brain barrier (BBB) contributes to Abeta removal from the brain. P-glycoprotein (Pgp) is highly expressed on the luminal surface of brain capillary endothelial cells and contributes to the BBB. In Pgp-null mice, we show that [I]Abeta40 and [I]Abeta42 microinjected into the CNS clear at half the rate that they do in WT mice. When amyloid precursor protein-transgenic (APP-transgenic) mice were administered a Pgp inhibitor, Abeta levels within the brain interstitial fluid significantly increased within hours of treatment. Furthermore, APP-transgenic, Pgp-null mice had increased levels of brain Abeta and enhanced Abeta deposition compared with APP-transgenic, Pgp WT mice. These data establish a direct link between Pgp and Abeta metabolism in vivo and suggest that Pgp activity at the BBB could affect risk for developing AD as well as provide a novel diagnostic and therapeutic target.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/deficiency , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Disease Models, Animal , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Polymorphism, Genetic , Up-Regulation/genetics
15.
J Clin Invest ; 128(5): 2144-2155, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29600961

ABSTRACT

The apolipoprotein E E4 allele of the APOE gene is the strongest genetic factor for late-onset Alzheimer disease (LOAD). There is compelling evidence that apoE influences Alzheimer disease (AD) in large part by affecting amyloid ß (Aß) aggregation and clearance; however, the molecular mechanism underlying these findings remains largely unknown. Herein, we tested whether anti-human apoE antibodies can decrease Aß pathology in mice producing both human Aß and apoE4, and investigated the mechanism underlying these effects. We utilized APPPS1-21 mice crossed to apoE4-knockin mice expressing human apoE4 (APPPS1-21/APOE4). We discovered an anti-human apoE antibody, anti-human apoE 4 (HAE-4), that specifically recognizes human apoE4 and apoE3 and preferentially binds nonlipidated, aggregated apoE over the lipidated apoE found in circulation. HAE-4 also binds to apoE in amyloid plaques in unfixed brain sections and in living APPPS1-21/APOE4 mice. When delivered centrally or by peripheral injection, HAE-4 reduced Aß deposition in APPPS1-21/APOE4 mice. Using adeno-associated virus to express 2 different full-length anti-apoE antibodies in the brain, we found that HAE antibodies decreased amyloid accumulation, which was dependent on Fcγ receptor function. These data support the hypothesis that a primary mechanism for apoE-mediated plaque formation may be a result of apoE aggregation, as preferentially targeting apoE aggregates with therapeutic antibodies reduces Aß pathology and may represent a selective approach to treat AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal, Murine-Derived/pharmacology , Apolipoprotein E4/antagonists & inhibitors , Plaque, Amyloid/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , Apolipoprotein E3/antagonists & inhibitors , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Humans , Mice , Mice, Knockout , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
16.
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
17.
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
18.
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
19.
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
20.
Neurobiol Dis ; 24(3): 506-15, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17010630

ABSTRACT

Although there are no proven ways to delay onset or slow progression of Alzheimer's disease (AD), studies suggest that diet can affect risk. Pomegranates contain very high levels of antioxidant polyphenolic substances as compared to other fruits and vegetables. Polyphenols have been shown to be neuroprotective in different model systems. We asked whether dietary supplementation with pomegranate juice (PJ) would influence behavior and AD-like pathology in a transgenic mouse model. Transgenic mice (APP(sw)/Tg2576) received either PJ or sugar water control from 6 to 12.5 months of age. PJ-treated mice learned water maze tasks more quickly and swam faster than controls. Mice treated with PJ had significantly less (approximately 50%) accumulation of soluble Abeta42 and amyloid deposition in the hippocampus as compared to control mice. These results suggest that further studies to validate and determine the mechanism of these effects, as well as whether substances in PJ may be useful in AD, should be considered.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Beverages , Hippocampus/metabolism , Lythraceae , Maze Learning/physiology , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/drug effects , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Antioxidants/administration & dosage , Dietary Supplements , Disease Models, Animal , Flavonoids/administration & dosage , Hippocampus/drug effects , Maze Learning/drug effects , Mice , Mice, Transgenic , Phenols/administration & dosage , Plaque, Amyloid/drug effects , Polyphenols , Spatial Behavior/drug effects , Spatial Behavior/physiology
SELECTION OF CITATIONS
SEARCH DETAIL