Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 150
Filter
Add more filters

Publication year range
1.
Cell ; 142(3): 387-97, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20655099

ABSTRACT

Alzheimer's disease (AD) is characterized by amyloid-beta (Abeta) and tau deposition in brain. It has emerged that Abeta toxicity is tau dependent, although mechanistically this link remains unclear. Here, we show that tau, known as axonal protein, has a dendritic function in postsynaptic targeting of the Src kinase Fyn, a substrate of which is the NMDA receptor (NR). Missorting of tau in transgenic mice expressing truncated tau (Deltatau) and absence of tau in tau(-/-) mice both disrupt postsynaptic targeting of Fyn. This uncouples NR-mediated excitotoxicity and hence mitigates Abeta toxicity. Deltatau expression and tau deficiency prevent memory deficits and improve survival in Abeta-forming APP23 mice, a model of AD. These deficits are also fully rescued with a peptide that uncouples the Fyn-mediated interaction of NR and PSD-95 in vivo. Our findings suggest that this dendritic role of tau confers Abeta toxicity at the postsynapse with direct implications for pathogenesis and treatment of AD.


Subject(s)
Alzheimer Disease/physiopathology , Dendrites/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Animals , Brain/pathology , Disks Large Homolog 4 Protein , Guanylate Kinases , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Memory Disorders/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-fyn/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , tau Proteins/genetics
2.
Proc Natl Acad Sci U S A ; 119(24): e2119804119, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35666874

ABSTRACT

Single-cell transcriptomics has revealed specific glial activation states associated with the pathogenesis of neurodegenerative diseases, such as Alzheimer's and Parkinson's disease. While these findings may eventually lead to new therapeutic opportunities, little is known about how these glial responses are reflected by biomarker changes in bodily fluids. Such knowledge, however, appears crucial for patient stratification, as well as monitoring disease progression and treatment responses in clinical trials. Here, we took advantage of well-described mouse models of ß-amyloidosis and α-synucleinopathy to explore cerebrospinal fluid (CSF) proteome changes related to their respective proteopathic lesions. Nontargeted liquid chromatography-mass spectrometry revealed that the majority of proteins that undergo age-related changes in CSF of either mouse model were linked to microglia and astrocytes. Specifically, we identified a panel of more than 20 glial-derived proteins that were increased in CSF of aged ß-amyloid precursor protein- and α-synuclein-transgenic mice and largely overlap with previously described disease-associated glial genes identified by single-cell transcriptomics. Our results also show that enhanced shedding is responsible for the increase of several of the identified glial CSF proteins as exemplified for TREM2. Notably, the vast majority of these proteins can also be quantified in human CSF and reveal changes in Alzheimer's disease cohorts. The finding that cellular transcriptome changes translate into corresponding changes of CSF proteins is of clinical relevance, supporting efforts to identify fluid biomarkers that reflect the various functional states of glial responses in cerebral proteopathies, such as Alzheimer's and Parkinson's disease.


Subject(s)
Alzheimer Disease , Cerebrospinal Fluid , Neuroglia , Parkinson Disease , Proteome , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/metabolism , Animals , Biomarkers/cerebrospinal fluid , Cerebrospinal Fluid/metabolism , Gene Expression Profiling , Humans , Mice , Neuroglia/metabolism , Parkinson Disease/cerebrospinal fluid , Parkinson Disease/metabolism , Proteome/metabolism , Single-Cell Analysis , tau Proteins
3.
Nature ; 556(7701): 332-338, 2018 04.
Article in English | MEDLINE | ID: mdl-29643512

ABSTRACT

Innate immune memory is a vital mechanism of myeloid cell plasticity that occurs in response to environmental stimuli and alters subsequent immune responses. Two types of immunological imprinting can be distinguished-training and tolerance. These are epigenetically mediated and enhance or suppress subsequent inflammation, respectively. Whether immune memory occurs in tissue-resident macrophages in vivo and how it may affect pathology remains largely unknown. Here we demonstrate that peripherally applied inflammatory stimuli induce acute immune training and tolerance in the brain and lead to differential epigenetic reprogramming of brain-resident macrophages (microglia) that persists for at least six months. Strikingly, in a mouse model of Alzheimer's pathology, immune training exacerbates cerebral ß-amyloidosis and immune tolerance alleviates it; similarly, peripheral immune stimulation modifies pathological features after stroke. Our results identify immune memory in the brain as an important modifier of neuropathology.


Subject(s)
Brain/immunology , Brain/pathology , Immunity, Innate , Immunologic Memory , Nervous System Diseases/immunology , Nervous System Diseases/pathology , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloidosis/immunology , Amyloidosis/pathology , Animals , Disease Models, Animal , Epigenesis, Genetic , Female , Gene Expression Regulation/immunology , Humans , Immune Tolerance , Inflammation/genetics , Inflammation/immunology , Male , Mice , Microglia/immunology , Microglia/metabolism , Stroke/immunology , Stroke/pathology
4.
Ann Neurol ; 86(4): 561-571, 2019 10.
Article in English | MEDLINE | ID: mdl-31359452

ABSTRACT

OBJECTIVE: Clinical trials targeting ß-amyloid peptides (Aß) for Alzheimer disease (AD) failed for arguable reasons that include selecting the wrong stages of AD pathophysiology or Aß being the wrong target. Targeting Aß to prevent cerebral amyloid angiopathy (CAA) has not been rigorously followed, although the causal role of Aß for CAA and related hemorrhages is undisputed. CAA occurs with normal aging and to various degrees in AD, where its impact and treatment is confounded by the presence of parenchymal Aß deposition. METHODS: APPDutch mice develop CAA in the absence of parenchymal amyloid, mimicking hereditary cerebral hemorrhage with amyloidosis Dutch type (HCHWA-D). Mice were treated with a ß-site amyloid precursor protein cleaving enzyme 1 (BACE1) inhibitor. We used 3-dimensional ultramicroscopy and immunoassays for visualizing CAA and assessing Aß in cerebrospinal fluid (CSF) and brain. RESULTS: CAA onset in mice was at 22 to 24 months, first in frontal leptomeningeal and superficial cortical vessels followed by vessels penetrating the cortical layers. CSF Aß increased with aging followed by a decrease of both Aß40 and Aß42 upon CAA onset, supporting the idea that combined reduction of CSF Aß40 and Aß42 is a specific biomarker for vascular amyloid. BACE1 inhibitor treatment starting at CAA onset and continuing for 4 months revealed a 90% Aß reduction in CSF and largely prevented CAA progression and associated pathologies. INTERPRETATION: This is the first study showing that Aß reduction at early disease time points largely prevents CAA in the absence of parenchymal amyloid. Our observation provides a preclinical basis for Aß-reducing treatments in patients at risk of CAA and in presymptomatic HCHWA-D. ANN NEUROL 2019;86:561-571.


Subject(s)
Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/metabolism , Brain/metabolism , Cerebral Amyloid Angiopathy/drug therapy , Disease Progression , Picolinic Acids/therapeutic use , Thiazines/therapeutic use , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Biomarkers/cerebrospinal fluid , Biomarkers/metabolism , Brain/blood supply , Female , Humans , Mice , Mice, Transgenic , Peptide Fragments/cerebrospinal fluid , Picolinic Acids/pharmacology , Thiazines/pharmacology
5.
Proc Natl Acad Sci U S A ; 114(32): 8631-8636, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28739891

ABSTRACT

Amyloid-ß (Aß) is thought to play an essential pathogenic role in Alzheimer´s disease (AD). A key enzyme involved in the generation of Aß is the ß-secretase BACE, for which powerful inhibitors have been developed and are currently in use in human clinical trials. However, although BACE inhibition can reduce cerebral Aß levels, whether it also can ameliorate neural circuit and memory impairments remains unclear. Using histochemistry, in vivo Ca2+ imaging, and behavioral analyses in a mouse model of AD, we demonstrate that along with reducing prefibrillary Aß surrounding plaques, the inhibition of BACE activity can rescue neuronal hyperactivity, impaired long-range circuit function, and memory defects. The functional neuronal impairments reappeared after infusion of soluble Aß, mechanistically linking Aß pathology to neuronal and cognitive dysfunction. These data highlight the potential benefits of BACE inhibition for the effective treatment of a wide range of AD-like pathophysiological and cognitive impairments.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Neurons/metabolism , Protease Inhibitors/pharmacology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/genetics , Animals , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Neurons/pathology
6.
Proc Natl Acad Sci U S A ; 114(49): 13018-13023, 2017 12 05.
Article in English | MEDLINE | ID: mdl-29158413

ABSTRACT

The molecular architecture of amyloids formed in vivo can be interrogated using luminescent conjugated oligothiophenes (LCOs), a unique class of amyloid dyes. When bound to amyloid, LCOs yield fluorescence emission spectra that reflect the 3D structure of the protein aggregates. Given that synthetic amyloid-ß peptide (Aß) has been shown to adopt distinct structural conformations with different biological activities, we asked whether Aß can assume structurally and functionally distinct conformations within the brain. To this end, we analyzed the LCO-stained cores of ß-amyloid plaques in postmortem tissue sections from frontal, temporal, and occipital neocortices in 40 cases of familial Alzheimer's disease (AD) or sporadic (idiopathic) AD (sAD). The spectral attributes of LCO-bound plaques varied markedly in the brain, but the mean spectral properties of the amyloid cores were generally similar in all three cortical regions of individual patients. Remarkably, the LCO amyloid spectra differed significantly among some of the familial and sAD subtypes, and between typical patients with sAD and those with posterior cortical atrophy AD. Neither the amount of Aß nor its protease resistance correlated with LCO spectral properties. LCO spectral amyloid phenotypes could be partially conveyed to Aß plaques induced by experimental transmission in a mouse model. These findings indicate that polymorphic Aß-amyloid deposits within the brain cluster as clouds of conformational variants in different AD cases. Heterogeneity in the molecular architecture of pathogenic Aß among individuals and in etiologically distinct subtypes of AD justifies further studies to assess putative links between Aß conformation and clinical phenotype.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Amyloid/chemistry , Plaque, Amyloid/metabolism , Protein Aggregates , Alzheimer Disease/classification , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid/classification , Amyloid/ultrastructure , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Female , Fluorescent Dyes/chemistry , Frontal Lobe/chemistry , Frontal Lobe/metabolism , Frontal Lobe/pathology , Gene Expression , Humans , Male , Mice , Occipital Lobe/chemistry , Occipital Lobe/metabolism , Occipital Lobe/pathology , Peptide Hydrolases/chemistry , Plaque, Amyloid/classification , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology , Presenilin-1/genetics , Presenilin-1/metabolism , Protein Binding , Protein Conformation , Proteolysis , Spectrometry, Fluorescence , Temporal Lobe/chemistry , Temporal Lobe/metabolism , Temporal Lobe/pathology , Thiophenes/chemistry
7.
Acta Neuropathol ; 138(6): 913-941, 2019 12.
Article in English | MEDLINE | ID: mdl-31414210

ABSTRACT

Extracellular deposition of amyloid ß-protein (Aß) in amyloid plaques and intracellular accumulation of abnormally phosphorylated τ-protein (p-τ) in neurofibrillary tangles (NFTs) represent pathological hallmark lesions of Alzheimer's disease (AD). Both lesions develop in parallel in the human brain throughout the preclinical and clinical course of AD. Nevertheless, it is not yet clear whether there is a direct link between Aß and τ pathology or whether other proteins are involved in this process. To address this question, we crossed amyloid precursor protein (APP) transgenic mice overexpressing human APP with the Swedish mutation (670/671 KM → NL) (APP23), human wild-type APP (APP51/16), or a proenkephalin signal peptide linked to human Aß42 (APP48) with τ-transgenic mice overexpressing human mutant 4-repeat τ-protein with the P301S mutation (TAU58). In 6-month-old APP23xTAU58 and APP51/16xTAU58 mice, soluble Aß was associated with the aggravation of p-τ pathology propagation into the CA1/subiculum region, whereas 6-month-old TAU58 and APP48xTAU58 mice neither exhibited significant amounts of p-τ pathology in the CA1/subiculum region nor displayed significant levels of soluble Aß in the forebrain. In APP23xTAU58 and APP51/16xTAU58 mice showing an acceleration of p-τ propagation, Aß and p-τ were co-immunoprecipitated with cellular prion protein (PrPC). A similar interaction between PrPC, p-τ and Aß was observed in human AD brains. This association was particularly noticed in 60% of the symptomatic AD cases in our sample, suggesting that PrPC may play a role in the progression of AD pathology. An in vitro pull-down assay confirmed that PrPC is capable of interacting with Aß and p-τ. Using a proximity ligation assay, we could demonstrate proximity (less than ~ 30-40 nm distance) between PrPC and Aß and between PrPC and p-τ in APP23xTAU58 mouse brain as well as in human AD brain. Proximity between PrPC and p-τ was also seen in APP51/16xTAU58, APP48xTAU58, and TAU58 mice. Based on these findings, it is tempting to speculate that PrPC is a critical player in the interplay between Aß and p-τ propagation at least in a large group of AD cases. Preexisting p-τ pathology interacting with PrPC, thereby, appears to be a prerequisite for Aß to function as a p-τ pathology accelerator via PrPC.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Neurofibrillary Tangles/pathology , tau Proteins/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Humans , Mice, Transgenic , Mutation/genetics , Plaque, Amyloid/pathology , Prion Proteins/metabolism
8.
EMBO Rep ; 18(9): 1536-1544, 2017 09.
Article in English | MEDLINE | ID: mdl-28701326

ABSTRACT

Little is known about the extent to which pathogenic factors drive the development of Alzheimer's disease (AD) at different stages of the long preclinical and clinical phases. Given that the aggregation of the ß-amyloid peptide (Aß) is an important factor in AD pathogenesis, we asked whether Aß seeds from brain extracts of mice at different stages of amyloid deposition differ in their biological activity. Specifically, we assessed the effect of age on Aß seeding activity in two mouse models of cerebral Aß amyloidosis (APPPS1 and APP23) with different ages of onset and rates of progression of Aß deposition. Brain extracts from these mice were serially diluted and inoculated into host mice. Strikingly, the seeding activity (seeding dose SD50) in extracts from donor mice of both models reached a plateau relatively early in the amyloidogenic process. When normalized to total brain Aß, the resulting specific seeding activity sharply peaked at the initial phase of Aß deposition, which in turn is characterized by a temporary several-fold increase in the Aß42/Aß40 ratio. At all stages, the specific seeding activity of the APPPS1 extract was higher compared to that of APP23 brain extract, consistent with a more important contribution of Aß42 than Aß40 to seed activity. Our findings indicate that the Aß seeding potency is greatest early in the pathogenic cascade and diminishes as Aß increasingly accumulates in brain. The present results provide experimental support for directing anti-Aß therapeutics to the earliest stage of the pathogenic cascade, preferably before the onset of amyloid deposition.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloidosis/metabolism , Brain/metabolism , Age Factors , Alzheimer Disease/drug therapy , Amyloidosis/drug therapy , Amyloidosis/physiopathology , Animals , Brain/pathology , Disease Models, Animal , Disease Progression , Humans , Mice , Mice, Transgenic
9.
Bioorg Med Chem Lett ; 28(12): 2195-2200, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29764741

ABSTRACT

New amino-1,4-oxazine derived BACE-1 inhibitors were explored and various synthetic routes developed. The binding mode of the inhibitors was elucidated by co-crystallization of 4 with BACE-1 and X-ray analysis. Subsequent optimization led to inhibitors with low double digit nanomolar activity in a biochemical and single digit nanomolar potency in a cellular assays. To assess the inhibitors for their permeation properties and potential to cross the blood-brain-barrier a MDR1-MDCK cell model was successfully applied. Compound 8a confirmed the in vitro results by dose-dependently reducing Aß levels in mice in an acute treatment regimen.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/pharmacology , Oxazines/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Dogs , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Madin Darby Canine Kidney Cells/drug effects , Mice , Models, Molecular , Molecular Conformation , Oxazines/chemical synthesis , Oxazines/chemistry , Structure-Activity Relationship
10.
J Neurosci ; 36(18): 5084-93, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27147660

ABSTRACT

UNLABELLED: The aggregation of amyloid-ß peptide (Aß) in brain is an early event and hallmark of Alzheimer's disease (AD). We combined the advantages of in vitro and in vivo approaches to study cerebral ß-amyloidosis by establishing a long-term hippocampal slice culture (HSC) model. While no Aß deposition was noted in untreated HSCs of postnatal Aß precursor protein transgenic (APP tg) mice, Aß deposition emerged in HSCs when cultures were treated once with brain extract from aged APP tg mice and the culture medium was continuously supplemented with synthetic Aß. Seeded Aß deposition was also observed under the same conditions in HSCs derived from wild-type or App-null mice but in no comparable way when HSCs were fixed before cultivation. Both the nature of the brain extract and the synthetic Aß species determined the conformational characteristics of HSC Aß deposition. HSC Aß deposits induced a microglia response, spine loss, and neuritic dystrophy but no obvious neuron loss. Remarkably, in contrast to in vitro aggregated synthetic Aß, homogenates of Aß deposits containing HSCs induced cerebral ß-amyloidosis upon intracerebral inoculation into young APP tg mice. Our results demonstrate that a living cellular environment promotes the seeded conversion of synthetic Aß into a potent in vivo seeding-active form. SIGNIFICANCE STATEMENT: In this study, we report the seeded induction of Aß aggregation and deposition in long-term hippocampal slice cultures. Remarkably, we find that the biological activities of the largely synthetic Aß aggregates in the culture are very similar to those observed in vivo This observation is the first to show that potent in vivo seeding-active Aß aggregates can be obtained by seeded conversion of synthetic Aß in a living (wild-type) cellular environment.


Subject(s)
Amyloid beta-Peptides/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/pathology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Neurites/pathology , Neurons/pathology , Organ Culture Techniques
11.
Alzheimers Dement ; 13(6): 701-709, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27750032

ABSTRACT

INTRODUCTION: The inhibition of the ß-site amyloid precursor protein-cleaving enzyme 1 (BACE1) is a main therapeutic approach for the treatment of Alzheimer's disease (AD). We previously reported an age-related increase of tau protein in the cerebrospinal fluid (CSF) of amyloid ß (Aß) precursor protein (APP) transgenic mice. METHODS: APP transgenic mice were treated with a potent BACE1 inhibitor. CSF tau and CSF Aß levels were assessed. A novel high-sensitivity tau sandwich immunoassay was developed. RESULTS: We demonstrate that long-term BACE1 inhibition prevents CSF tau increase both in early-depositing APP transgenic mice and APP transgenic mice with moderate Aß pathology. DISCUSSION: Our results demonstrate that BACE1 inhibition not only reduces Aß generation but also downstream AD pathophysiology. The tight correlation between Aß aggregation in brain and CSF tau levels renders CSF tau a valuable marker to predict the effectiveness of BACE1 inhibitors in current clinical trials.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/cerebrospinal fluid , Aspartic Acid Endopeptidases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Picolinic Acids/pharmacology , Thiazines/pharmacology , tau Proteins/cerebrospinal fluid , Aging/drug effects , Aging/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Female , Humans , Immunoassay , Male , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid/drug therapy , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Prosencephalon/drug effects , Prosencephalon/metabolism , Prosencephalon/pathology
12.
Hum Mol Genet ; 23(24): 6512-27, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25027328

ABSTRACT

Normally, sonic hedgehog (Shh) signaling induces high levels of Patched 1 (Ptc1) and its associated transcription factor Gli1 with genesis of specific neuronal progeny. But their roles in the neural stem cells (NSCs), including glial precursor cells (GPCs), of Alzheimer's disease (AD) are unclear. Here, we show that Ptc1 and Gli1 are significantly deficits in the hippocampus of an aged AD transgenic mouse mode, whereas these two molecules are highly elevated at young ages. Our similar findings in autopsied AD brains validate the discovery in AD mouse models. To examine whether Aß peptides, which are a main component of the amyloid plaques in AD brains, affected Ptc1-Gli1 signaling, we treated GPCs with Aß peptides, we found that high dose of Aß1-42 but not Aß1-40 significantly decreased Ptc1-Gli1, while Shh itself was elevated in hippocampal NSCs/GPCs. Furthermore, we found that deficits of Ptc1-Gli1 signaling induced NSCs/GPCs into asymmetric division, which results in an increase in the number of dividing cells including transit-amplifying cells and neuroblasts. These precursor cells commit to apoptosis-like death under the toxic conditions. By this way, adult neural precursor cell pool is exhausted and defective neurogenesis happens in AD brains. Our findings suggest that Ptc1-Gli1 signaling deregulation resulting abnormal loss of GPCs may contribute to a cognition decline in AD brains. The novel findings elucidate a new molecular mechanism of adult NSCs/GPCs on neurogenesis and demonstrate a regulatory role for Ptc1-Gli1 in adult neural circuit integrity of the brain.


Subject(s)
Alzheimer Disease/genetics , Neural Stem Cells/metabolism , Neuroglia/metabolism , Receptors, Cell Surface/genetics , Signal Transduction , Transcription Factors/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Animals , Apoptosis/drug effects , Cell Division/drug effects , Disease Models, Animal , Female , Gene Expression Regulation , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Transgenic , Neural Stem Cells/drug effects , Neural Stem Cells/pathology , Neurogenesis/genetics , Neuroglia/drug effects , Neuroglia/pathology , Patched Receptors , Patched-1 Receptor , Peptide Fragments/pharmacology , Primary Cell Culture , Receptors, Cell Surface/metabolism , Transcription Factors/metabolism , Zinc Finger Protein GLI1
13.
Hum Mol Genet ; 23(18): 4906-18, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24824215

ABSTRACT

Tumor necrosis factor receptor II (TNFRII) is one of the TNF receptor superfamily members and our recent pathological studies show that TNFRII is deficient in the brains of Alzheimer's disease (AD). However, the mechanisms of TNFRII in AD pathogenesis remain unclear. In the present study, by using the gene-targeting approach to delete TNFRII in AD transgenic mouse model, we found that, in the brain of APP23 mice with TNFRII deletion (APP23/TNFRII(-/-)), AD-like pathology, i.e. plaque formation and microglial activation, occurs as early as 6 months of age. To test whether the increased levels of Aß plaques was due to elevated Aß, we measured Aß and found that Aß levels indeed were significantly increased at this age. Because ß-secretase, BACE1, is critical enzyme for Aß production, we have examined BACE1 and found that BACE1 is increased in both protein levels and enzymatic activity as early as 6 months of age; Having shown that BACE1 promoter region contains NF-κB binding sites, we found that cytoplasmic NF-κB was elevated and SUMO1 binding to IκBα was decreased. To further verify these findings, we have overexpressed TNFRII and identified that overexpressing TNFRII can reverse the findings from APP23/TNFRII(-/-) mice. Altogether, our results demonstrate novel roles of TNFRII in the regulation of Aß production, suggesting a potential therapeutic strategy for AD by up-regulating TNFRII levels and elevating phosphorylated IκBα by SUMOylation.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , I-kappa B Proteins/metabolism , Receptors, Tumor Necrosis Factor, Type II/genetics , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Binding Sites , Disease Models, Animal , Gene Deletion , Gene Targeting/methods , Humans , Mice , Mice, Transgenic , Microglia/metabolism , NF-KappaB Inhibitor alpha , Phosphorylation , Receptors, Tumor Necrosis Factor, Type II/metabolism
14.
Neurobiol Learn Mem ; 130: 34-43, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26827932

ABSTRACT

Frontotemporal lobar degeneration (FTLD) is a neurodegenerative disorder, a major subset of which is characterized by the accumulation of abnormal forms of the protein tau, leading to impairments in motor functions as well as language and behavioral alterations. Tau58-2/B mice express human tau with the P301S mutation found in familial forms of FTLD in neurons. By assessing three age cohorts of Tau58-2/B mice in a comprehensive behavioral test battery, we found that the tauopathy animals showed age-dependent signs of impulsivity, decreased social exploration and executive dysfunction. The deficit in executive function was first limited to decreased spatial working memory, but with aging this was extended to impaired instrumental short-term memory. Tau pathology was prominent in brain regions underlying these behaviors. Thus, Tau-58-2/B mice recapitulate neurological deficits of the behavioral variant of frontotemporal dementia (bvFTD), presenting them as a suitable model to test therapeutic interventions for the amelioration of this variant.


Subject(s)
Behavior, Animal/physiology , Executive Function/physiology , Exploratory Behavior/physiology , Frontotemporal Dementia/psychology , Impulsive Behavior/physiology , Social Behavior , Animals , Cerebral Cortex/pathology , Disease Models, Animal , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Hippocampus/pathology , Mice , Mutation , tau Proteins/genetics
15.
Proc Natl Acad Sci U S A ; 110(23): 9535-40, 2013 Jun 04.
Article in English | MEDLINE | ID: mdl-23690619

ABSTRACT

Filamentous inclusions made of hyperphosphorylated tau are characteristic of numerous human neurodegenerative diseases, including Alzheimer's disease, tangle-only dementia, Pick disease, argyrophilic grain disease (AGD), progressive supranuclear palsy, and corticobasal degeneration. In Alzheimer's disease and AGD, it has been shown that filamentous tau appears to spread in a stereotypic manner as the disease progresses. We previously demonstrated that the injection of brain extracts from human mutant P301S tau-expressing transgenic mice into the brains of mice transgenic for wild-type human tau (line ALZ17) resulted in the assembly of wild-type human tau into filaments and the spreading of tau inclusions from the injection sites to anatomically connected brain regions. Here we injected brain extracts from humans who had died with various tauopathies into the hippocampus and cerebral cortex of ALZ17 mice. Argyrophilic tau inclusions formed in all cases and following the injection of the corresponding brain extracts, we recapitulated the hallmark lesions of AGD, PSP and CBD. Similar inclusions also formed after intracerebral injection of brain homogenates from human tauopathies into nontransgenic mice. Moreover, the induced formation of tau aggregates could be propagated between mouse brains. These findings suggest that once tau aggregates have formed in discrete brain areas, they become self-propagating and spread in a prion-like manner.


Subject(s)
Brain/metabolism , Tauopathies/physiopathology , Tissue Extracts/pharmacology , tau Proteins/metabolism , Aged , Aged, 80 and over , Animals , Blotting, Western , Brain/pathology , Crosses, Genetic , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tissue Extracts/administration & dosage , Transplantation, Heterologous , tau Proteins/genetics
16.
Int J Mol Sci ; 17(1)2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26797603

ABSTRACT

BACKGROUND: Cerebral amyloid angiopathy (CAA) is characterized by vascular deposition of amyloid ß (Aß) with a higher incidence of cerebral microbleeds (cMBs) and spontaneous hemorrhage. Since statins are known for their benefit in vascular disease we tested for the effect on CAA. METHODS: APP23-transgenic mice received atorvastatin-supplemented food starting at the age of eight months (n = 13), 12 months (n = 7), and 16 months (n = 6), respectively. Controls (n = 16) received standard food only. At 24 months of age cMBs were determined with T2*-weighted 9.4T magnetic resonance imaging and graded by size. RESULTS: Control mice displayed an average of 35 ± 18.5 cMBs (mean ± standard deviation), compared to 29.3 ± 9.8 in mice with eight months (p = 0.49), 24.9 ± 21.3 with 12 months (p = 0.26), and 27.8 ± 15.4 with 16 months of atorvastatin treatment (p = 0.27). In combined analysis treated mice showed lower absolute numbers (27.4 ± 15.6, p = 0.16) compared to controls and also after adjustment for cMB size (p = 0.13). CONCLUSION: Despite to a non-significant trend towards fewer cMBs our results failed to provide evidence for beneficial effects of long-term atorvastatin treatment in the APP23-transgenic mouse model of CAA. A higher risk for bleeding complications was not observed.


Subject(s)
Anticholesteremic Agents/pharmacology , Atorvastatin/pharmacology , Cerebral Amyloid Angiopathy/drug therapy , Cerebral Hemorrhage/drug therapy , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cerebral Amyloid Angiopathy/genetics , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/pathology , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Disease Models, Animal , Gene Expression , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Mice, Transgenic
17.
J Neurosci ; 34(31): 10264-73, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-25080588

ABSTRACT

Deposition of aggregated amyloid-ß (Aß) peptide in brain is an early event and hallmark pathology of Alzheimer's disease and cerebral Aß angiopathy. Experimental evidence supports the concept that Aß multimers can act as seeds and structurally corrupt other Aß peptides by a self-propagating mechanism. Here we compare the induction of cerebral ß-amyloidosis by intraperitoneal applications of Aß-containing brain extracts in three Aß-precursor protein (APP) transgenic mouse lines that differ in levels of transgene expression in brain and periphery (APP23 mice, APP23 mice lacking murine APP, and R1.40 mice). Results revealed that beta-amyloidosis induction, which could be blocked with an anti-Aß antibody, was dependent on the amount of inoculated brain extract and on the level of APP/Aß expression in the brain but not in the periphery. The induced Aß deposits in brain occurred in a characteristic pattern consistent with the entry of Aß seeds at multiple brain locations. Intraperitoneally injected Aß could be detected in blood monocytes and some peripheral tissues (liver, spleen) up to 30 d after the injection but escaped histological and biochemical detection thereafter. These results suggest that intraperitoneally inoculated Aß seeds are transported from the periphery to the brain in which corruptive templating of host Aß occurs at multiple sites, most efficiently in regions with high availability of soluble Aß.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Amyloidosis , Cerebral Cortex/pathology , Amyloid beta-Peptides/immunology , Amyloid beta-Protein Precursor/genetics , Amyloidosis/chemically induced , Amyloidosis/genetics , Amyloidosis/pathology , Animals , Antibodies/pharmacology , Blood Cells/metabolism , Blood Cells/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Dose-Response Relationship, Drug , Drug Administration Routes , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Peritoneal Cavity/pathology , Plaque, Amyloid/pathology , Time Factors
18.
Neuropathol Appl Neurobiol ; 41(7): 906-25, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25763777

ABSTRACT

AIM: Tau becomes hyperphosphorylated in Alzheimer's disease (AD) and frontotemporal lobar degeneration (FTLD-tau), resulting in functional deficits of neurones, neurofibrillary tangle (NFT) formation and eventually dementia. Expression of mutant human tau in the brains of transgenic mice has produced different lines that recapitulate various aspects of FTLD-tau and AD. In this study, we characterized the novel P301S mutant tau transgenic mouse line, TAU58/2. METHODS: Both young and aged TAU58/2 mice underwent extensive motor testing, after which brain tissue was analysed with immunohistochemistry, silver staining, electron microscopy and Western blotting. Tissue from various FTLD subtypes and AD patients was also analysed for comparison. RESULTS: TAU58/2 mice presented with early-onset motor deficits, which became more pronounced with age. Throughout the brains of these mice, tau was progressively hyperphosphorylated resulting in increased NFT formation with age. In addition, frequent axonal swellings that stained intensively for neurofilament (NF) were present in young TAU58/2 mice prior to NFT formation. Similar axonal pathology was also observed in human FTLD-tau and AD. Interestingly, activated microglia were found in close proximity to neurones harbouring transgenic tau, but were not associated with NF-positive axonal swellings. CONCLUSIONS: In TAU58/2 mice, early tau pathology induces functional deficits of neurones associated with NF pathology. This appears to be specific to tau, as similar changes are observed in FTLD-tau, but not in FTLD with TDP-43 inclusions. Therefore, TAU58/2 mice recapitulate aspects of human FTLD-tau and AD pathology, and will become instrumental in studying disease mechanisms and therapeutics in the future.


Subject(s)
Axons/pathology , Brain/pathology , Frontotemporal Lobar Degeneration/pathology , Neurons/pathology , Animals , Brain/metabolism , Disease Models, Animal , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/metabolism , Mice , Mice, Transgenic , Neurons/metabolism , tau Proteins/genetics , tau Proteins/metabolism
19.
Brain ; 137(Pt 11): 2909-2915, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25212850

ABSTRACT

The soluble fraction of brain samples from patients with Alzheimer's disease contains highly biologically active amyloid-ß seeds. In this study, we sought to assess the potency of soluble amyloid-ß seeds derived from the brain and cerebrospinal fluid. Soluble Alzheimer's disease brain extracts were serially diluted and then injected into the hippocampus of young, APP transgenic mice. Eight months later, seeded amyloid-ß deposition was evident even when the hippocampus received subattomole amounts of brain-derived amyloid-ß. In contrast, cerebrospinal fluid from patients with Alzheimer's disease, which contained more than 10-fold higher levels of amyloid-ß peptide than the most concentrated soluble brain extracts, did not induce detectable seeding activity in vivo. Similarly, cerebrospinal fluid from aged APP-transgenic donor mice failed to induce cerebral amyloid-ß deposition. In comparison to the soluble brain fraction, cerebrospinal fluid largely lacked N-terminally truncated amyloid-ß species and exhibited smaller amyloid-ß-positive particles, features that may contribute to the lack of in vivo seeding by cerebrospinal fluid. Interestingly, the same cerebrospinal fluid showed at least some seeding activity in an in vitro assay. The present results indicate that the biological seeding activity of soluble amyloid-ß species is orders of magnitude greater in brain extracts than in the cerebrospinal fluid.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Frontal Lobe/metabolism , Hippocampus/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/chemistry , Amyloid beta-Protein Precursor/genetics , Animals , Female , Frontal Lobe/pathology , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Peptide Fragments/cerebrospinal fluid , Peptide Fragments/chemistry , Random Allocation , Solubility
20.
Proc Natl Acad Sci U S A ; 109(22): 8740-5, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22592800

ABSTRACT

Alzheimer's disease (AD) is characterized by a progressive dysfunction of central neurons. Recent experimental evidence indicates that in the cortex, in addition to the silencing of a fraction of neurons, other neurons are hyperactive in amyloid-ß (Aß) plaque-enriched regions. However, it has remained unknown what comes first, neuronal silencing or hyperactivity, and what mechanisms might underlie the primary neuronal dysfunction. Here we examine the activity patterns of hippocampal CA1 neurons in a mouse model of AD in vivo using two-photon Ca(2+) imaging. We found that neuronal activity in the plaque-bearing CA1 region of older mice is profoundly altered. There was a marked increase in the fractions of both silent and hyperactive neurons, as previously also found in the cortex. Remarkably, in the hippocampus of young mice, we observed a selective increase in hyperactive neurons already before the formation of plaques, suggesting that soluble species of Aß may underlie this impairment. Indeed, we found that acute treatment with the γ-secretase inhibitor LY-411575 reduces soluble Aß levels and rescues the neuronal dysfunction. Furthermore, we demonstrate that direct application of soluble Aß can induce neuronal hyperactivity in wild-type mice. Thus, our study identifies hippocampal hyperactivity as a very early functional impairment in AD transgenic mice and provides direct evidence that soluble Aß is crucial for hippocampal hyperactivity.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/physiology , Disease Models, Animal , Hippocampus/physiopathology , Age Factors , Alanine/analogs & derivatives , Alanine/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Animals , Azepines/pharmacology , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/physiopathology , Calcium/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Humans , Mice , Mice, Transgenic , Microscopy, Fluorescence, Multiphoton , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
SELECTION OF CITATIONS
SEARCH DETAIL