Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Nat Commun ; 13(1): 7333, 2022 11 28.
Article in English | MEDLINE | ID: mdl-36443293

ABSTRACT

Brain Aß deposition is a key early event in the pathogenesis of Alzheimer´s disease (AD), but the long presymptomatic phase and poor correlation between Aß deposition and clinical symptoms remain puzzling. To elucidate the dependency of downstream pathologies on Aß, we analyzed the trajectories of cerebral Aß accumulation, Aß seeding activity, and neurofilament light chain (NfL) in the CSF (a biomarker of neurodegeneration) in Aß-precursor protein transgenic mice. We find that Aß deposition increases linearly until it reaches an apparent plateau at a late age, while Aß seeding activity increases more rapidly and reaches a plateau earlier, coinciding with the onset of a robust increase of CSF NfL. Short-term inhibition of Aß generation in amyloid-laden mice reduced Aß deposition and associated glial changes, but failed to reduce Aß seeding activity, and CSF NfL continued to increase although at a slower pace. When short-term or long-term inhibition of Aß generation was started at pre-amyloid stages, CSF NfL did not increase despite some Aß deposition, microglial activation, and robust brain Aß seeding activity. A dissociation of Aß load and CSF NfL trajectories was also found in familial AD, consistent with the view that Aß aggregation is not kinetically coupled to neurotoxicity. Rather, neurodegeneration starts when Aß seeding activity is saturated and before Aß deposition reaches critical (half-maximal) levels, a phenomenon reminiscent of the two pathogenic phases in prion disease.


Subject(s)
Alzheimer Disease , Amyloidosis , Animals , Mice , Brain , Disease Progression , Amyloidogenic Proteins , Inhibition, Psychological , Mice, Transgenic
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
4.
BMC Cancer ; 21(1): 1108, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34654395

ABSTRACT

BACKGROUND: Diffuse gliomas are the most common malignant tumors of the central nervous system with poor treatment efficacy. Infiltration of immune cells into tumors during immunosurveillance is observed in multiple tumor entities and often associated with a favorable outcome. The aim of this study was to evaluate the infiltration of immune cells in gliomas and their association with cerebrospinal fluid (CSF) cytokine concentrations. METHODS: We applied immunohistochemistry in tumor tissue sections of 18 high-grade glioma (HGG) patients (4 anaplastic astrocytoma, IDH-wildtype WHO-III; 14 glioblastomas (GBM), IDH-wildtype WHO-IV) in order to assess and quantify leucocytes (CD45) and macrophages (CD68, CD163) within the tumor core, infiltration zone and perivascular spaces. In addition, we quantified the concentrations of 30 cytokines in the same patients' CSF and in 14 non-inflammatory controls. RESULTS: We observed a significantly higher percentage of CD68+ macrophages (21-27%) in all examined tumor areas when compared to CD45+ leucocytes (ca. 3-7%); CD163+ cell infiltration was between 5 and 15%. Compared to the tumor core, significantly more macrophages and leucocytes were detectable within the perivascular area. The brain parenchyma showing a lower tumor cell density seems to be less infiltrated by macrophages. Interleukin (IL)-7 was significantly downregulated in CSF of GBM patients compared to controls. Additionally, CD68+ macrophage infiltrates showed significant correlations with the expression of eotaxin, interferon-γ, IL-1ß, IL-2, IL-10, IL-13, IL-16 and vascular endothelial growth factor. CONCLUSIONS: Our findings suggest that the infiltration of lymphocytes is generally low in HGG, and does not correlate with cytokine concentrations in the CSF. In contrast, macrophage infiltrates in HGG are associated with CSF cytokine changes that possibly shape the tumor microenvironment. Although results point towards an escape from immunosurveillance or even exploitation of immune cells by HGG, further studies are necessary to decipher the exact role of the immune system in these tumors.


Subject(s)
Astrocytoma/cerebrospinal fluid , Brain Neoplasms/cerebrospinal fluid , Cytokines/cerebrospinal fluid , Glioblastoma/cerebrospinal fluid , Leukocytes , Macrophages , Adult , Aged , Aged, 80 and over , Antigens, CD , Antigens, Differentiation, Myelomonocytic , Astrocytoma/pathology , Brain Neoplasms/pathology , Cell Count , Chemokine CCL11/cerebrospinal fluid , Female , Glioblastoma/pathology , Humans , Immunohistochemistry , Interferon-gamma/cerebrospinal fluid , Interleukins/cerebrospinal fluid , Leukocytes/cytology , Lymphocytes, Tumor-Infiltrating/cytology , Macrophages/cytology , Male , Middle Aged , Receptors, Cell Surface , Tumor Microenvironment , Vascular Endothelial Growth Factor A/cerebrospinal fluid
5.
Nat Aging ; 1(2): 218-225, 2021 02.
Article in English | MEDLINE | ID: mdl-37118632

ABSTRACT

Neurofilament light chain (NfL) has emerged as a promising blood biomarker for the progression of various neurological diseases. NfL is a structural protein of nerve cells, and elevated NfL levels in blood are thought to mirror damage to the nervous system. We find that plasma NfL levels increase in humans with age (n = 122; 21-107 years of age) and correlate with changes in other plasma proteins linked to neural pathways. In centenarians (n = 135), plasma NfL levels are associated with mortality equally or better than previously described multi-item scales of cognitive or physical functioning, and this observation was replicated in an independent cohort of nonagenarians (n = 180). Plasma NfL levels also increase in aging mice (n = 114; 2-30 months of age), and dietary restriction, a paradigm that extends lifespan in mice, attenuates the age-related increase in plasma NfL levels. These observations suggest a contribution of nervous system functional deterioration to late-life mortality.


Subject(s)
Aging , Neurons , Aged, 80 and over , Animals , Humans , Mice , Neurofilament Proteins/blood , Mortality
6.
Mol Psychiatry ; 26(10): 5733-5750, 2021 10.
Article in English | MEDLINE | ID: mdl-32632204

ABSTRACT

Mutations in pitrilysin metallopeptidase 1 (PITRM1), a mitochondrial protease involved in mitochondrial precursor processing and degradation, result in a slow-progressing syndrome characterized by cerebellar ataxia, psychotic episodes, and obsessive behavior, as well as cognitive decline. To investigate the pathogenetic mechanisms of mitochondrial presequence processing, we employed cortical neurons and cerebral organoids generated from PITRM1-knockout human induced pluripotent stem cells (iPSCs). PITRM1 deficiency strongly induced mitochondrial unfolded protein response (UPRmt) and enhanced mitochondrial clearance in iPSC-derived neurons. Furthermore, we observed increased levels of amyloid precursor protein and amyloid ß in PITRM1-knockout neurons. However, neither cell death nor protein aggregates were observed in 2D iPSC-derived cortical neuronal cultures. On the other hand, over time, cerebral organoids generated from PITRM1-knockout iPSCs spontaneously developed pathological features of Alzheimer's disease (AD), including the accumulation of protein aggregates, tau pathology, and neuronal cell death. Single-cell RNA sequencing revealed a perturbation of mitochondrial function in all cell types in PITRM1-knockout cerebral organoids, whereas immune transcriptional signatures were substantially dysregulated in astrocytes. Importantly, we provide evidence of a protective role of UPRmt and mitochondrial clearance against impaired mitochondrial presequence processing and proteotoxic stress. Here, we propose a novel concept of PITRM1-linked neurological syndrome whereby defects of mitochondrial presequence processing induce an early activation of UPRmt that, in turn, modulates cytosolic quality control pathways. Thus, our work supports a mechanistic link between mitochondrial function and common neurodegenerative proteinopathies.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Alzheimer Disease/genetics , Amyloid beta-Peptides , Humans , Metalloendopeptidases , Mitochondria , Organoids
7.
Nat Neurosci ; 23(12): 1580-1588, 2020 12.
Article in English | MEDLINE | ID: mdl-33199898

ABSTRACT

Amyloid-ß (Aß) deposits are a relatively late consequence of Aß aggregation in Alzheimer's disease. When pathogenic Aß seeds begin to form, propagate and spread is not known, nor are they biochemically defined. We tested various antibodies for their ability to neutralize Aß seeds before Aß deposition becomes detectable in Aß precursor protein-transgenic mice. We also characterized the different antibody recognition profiles using immunoprecipitation of size-fractionated, native, mouse and human brain-derived Aß assemblies. At least one antibody, aducanumab, after acute administration at the pre-amyloid stage, led to a significant reduction of Aß deposition and downstream pathologies 6 months later. This demonstrates that therapeutically targetable pathogenic Aß seeds already exist during the lag phase of protein aggregation in the brain. Thus, the preclinical phase of Alzheimer's disease-currently defined as Aß deposition without clinical symptoms-may be a relatively late manifestation of a much earlier pathogenic seed formation and propagation that currently escapes detection in vivo.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/antagonists & inhibitors , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacology , Brain Chemistry , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Neurofilament Proteins/cerebrospinal fluid , Plaque, Amyloid/pathology , Tissue Extracts/pharmacology
8.
EMBO Mol Med ; 12(7): e11803, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32510847

ABSTRACT

With molecular treatments coming into reach for spinocerebellar ataxia type 3 (SCA3), easily accessible, cross-species validated biomarkers for human and preclinical trials are warranted, particularly for the preataxic disease stage. We assessed serum levels of neurofilament light (NfL) and phosphorylated neurofilament heavy (pNfH) in ataxic and preataxic subjects of two independent multicentric SCA3 cohorts and in a SCA3 knock-in mouse model. Ataxic SCA3 subjects showed increased levels of both NfL and pNfH. In preataxic subjects, NfL levels increased with proximity to the individual expected onset of ataxia, with significant NfL elevations already 7.5 years before onset. Cross-sectional NfL levels correlated with both disease severity and longitudinal disease progression. Blood NfL and pNfH increases in human SCA3 were each paralleled by similar changes in SCA3 knock-in mice, here also starting already at the presymptomatic stage, closely following ataxin-3 aggregation and preceding Purkinje cell loss in the brain. Blood neurofilaments, particularly NfL, might thus provide easily accessible, cross-species validated biomarkers in both ataxic and preataxic SCA3, associated with earliest neuropathological changes, and serve as progression, proximity-to-onset and, potentially, treatment-response markers in both human and preclinical SCA3 trials.


Subject(s)
Intermediate Filaments , Machado-Joseph Disease/blood , Prodromal Symptoms , Animals , Biomarkers/blood , Cross-Sectional Studies , Female , Humans , Intermediate Filaments/chemistry , Male , Mice , Severity of Illness Index
9.
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
10.
Nat Med ; 25(2): 277-283, 2019 02.
Article in English | MEDLINE | ID: mdl-30664784

ABSTRACT

Neurofilament light chain (NfL) is a promising fluid biomarker of disease progression for various cerebral proteopathies. Here we leverage the unique characteristics of the Dominantly Inherited Alzheimer Network and ultrasensitive immunoassay technology to demonstrate that NfL levels in the cerebrospinal fluid (n = 187) and serum (n = 405) are correlated with one another and are elevated at the presymptomatic stages of familial Alzheimer's disease. Longitudinal, within-person analysis of serum NfL dynamics (n = 196) confirmed this elevation and further revealed that the rate of change of serum NfL could discriminate mutation carriers from non-mutation carriers almost a decade earlier than cross-sectional absolute NfL levels (that is, 16.2 versus 6.8 years before the estimated symptom onset). Serum NfL rate of change peaked in participants converting from the presymptomatic to the symptomatic stage and was associated with cortical thinning assessed by magnetic resonance imaging, but less so with amyloid-ß deposition or glucose metabolism (assessed by positron emission tomography). Serum NfL was predictive for both the rate of cortical thinning and cognitive changes assessed by the Mini-Mental State Examination and Logical Memory test. Thus, NfL dynamics in serum predict disease progression and brain neurodegeneration at the early presymptomatic stages of familial Alzheimer's disease, which supports its potential utility as a clinically useful biomarker.


Subject(s)
Alzheimer Disease/blood , Alzheimer Disease/pathology , Disease Progression , Nerve Degeneration/blood , Neurofilament Proteins/blood , Alzheimer Disease/cerebrospinal fluid , Humans , Mutation/genetics , Neurofilament Proteins/cerebrospinal fluid , Neurofilament Proteins/genetics
11.
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
12.
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
13.
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
15.
Neuron ; 91(1): 56-66, 2016 07 06.
Article in English | MEDLINE | ID: mdl-27292537

ABSTRACT

A majority of current disease-modifying therapeutic approaches for age-related neurodegenerative diseases target their characteristic proteopathic lesions (α-synuclein, Tau, Aß). To monitor such treatments, fluid biomarkers reflecting the underlying disease process are crucial. We found robust increases of neurofilament light chain (NfL) in CSF and blood in murine models of α-synucleinopathies, tauopathy, and ß-amyloidosis. Blood and CSF NfL levels were strongly correlated, and NfL increases coincided with the onset and progression of the corresponding proteopathic lesions in brain. Experimental induction of α-synuclein lesions increased CSF and blood NfL levels, while blocking Aß lesions attenuated the NfL increase. Consistently, we also found NfL increases in CSF and blood of human α-synucleinopathies, tauopathies, and Alzheimer's disease. Our results suggest that CSF and particularly blood NfL can serve as a reliable and easily accessible biomarker to monitor disease progression and treatment response in mouse models and potentially in human proteopathic neurodegenerative diseases.


Subject(s)
Intermediate Filaments/metabolism , Neurodegenerative Diseases/metabolism , Neurofilament Proteins/blood , Neurofilament Proteins/cerebrospinal fluid , Animals , Axons/metabolism , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Brain/metabolism , Brain/pathology , Disease Progression , Mice, Inbred C57BL , Mice, Transgenic , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/pathology , alpha-Synuclein/metabolism
16.
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
17.
Curr Alzheimer Res ; 13(6): 654-62, 2016.
Article in English | MEDLINE | ID: mdl-26971930

ABSTRACT

Altered progranulin levels play a major role in neurodegenerative diseases, like Alzheimer's dementia (AD), frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS), even in the absence of GRN mutations. Increasing progranulin levels could hereby provide a novel treatment strategy. However, knowledge on progranulin regulation in neurodegenerative diseases remains limited. We here demonstrate that cerebrospinal fluid progranulin levels do not correlate with its serum levels in AD, FTD and ALS, indicating a differential regulation of its central and peripheral levels in neurodegeneration. Blood progranulin levels thus do not reliably predict central nervous progranulin levels and their response to future progranulin-increasing therapeutics.


Subject(s)
Alzheimer Disease/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Frontotemporal Dementia/metabolism , Intercellular Signaling Peptides and Proteins/blood , Intercellular Signaling Peptides and Proteins/cerebrospinal fluid , Aged , Alzheimer Disease/genetics , Amyotrophic Lateral Sclerosis/genetics , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Female , Frontotemporal Dementia/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Middle Aged , Progranulins
18.
Nat Neurosci ; 18(11): 1559-61, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26352792

ABSTRACT

Cerebral ß-amyloidosis is induced by inoculation of Aß seeds into APP transgenic mice, but not into App(-/-) (APP null) mice. We found that brain extracts from APP null mice that had been inoculated with Aß seeds up to 6 months previously still induced ß-amyloidosis in APP transgenic hosts following secondary transmission. Thus, Aß seeds can persist in the brain for months, and they regain propagative and pathogenic activity in the presence of host Aß.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/pharmacology , Brain/drug effects , Brain/metabolism , Plaque, Amyloid/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/administration & dosage , Amyloid beta-Protein Precursor/deficiency , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/pathology , Animals , Brain/pathology , Disease Models, Animal , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Plaque, Amyloid/pathology
19.
Brain Res ; 1622: 149-62, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26115583

ABSTRACT

Hereditary Cystatin C Amyloid Angiopathy (HCCAA) is an amyloid disorder in Icelandic families caused by an autosomal dominant mutation in the cystatin C gene. Mutant cystatin C forms amyloid deposits in brain arteries and arterioles which are associated with changes in the arterial wall structure, notably deposition of extracellular matrix proteins. In this post-mortem study we examined the neuroinflammatory response relative to the topographical distribution of cystatin C deposition, and associated haemorrhages, in the leptomeninges, cerebrum, cerebellum, thalamus, and midbrain of HCCAA patients. Cystatin C was deposited in all brain areas, grey and white matter alike, most prominently in arteries and arterioles; capillaries and veins were not, or minimally, affected. We also observed perivascular deposits and parenchymal focal deposits proximal to affected arteries. This study shows for the first time, that cystatin C does not exclusively form CAA and perivascular amyloid but also focal deposits in the brain parenchyma. Haemorrhages were observed in all patients and occurred in all brain areas, variable between patients. Microinfarcts were observed in 34.6% of patients. The neuroinflammatory response was limited to the close vicinity of affected arteries and perivascular as well as parenchymal focal deposits. Taken together with previously reported arterial accumulation of extracellular matrix proteins in HCCAA, our results indicate that the central nervous system pathology of HCCAA is characterised by the formation of a glial scar within and around affected arteries.


Subject(s)
Brain/pathology , Cerebral Amyloid Angiopathy, Familial/pathology , Cicatrix/pathology , Cystatin C/metabolism , Neuroglia/pathology , Adult , Aged , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Arterioles/metabolism , Arterioles/pathology , Brain/blood supply , Brain/metabolism , Brain Infarction/pathology , Brain Infarction/physiopathology , Calcium-Binding Proteins , Cerebral Amyloid Angiopathy, Familial/genetics , Cerebral Amyloid Angiopathy, Familial/physiopathology , Cerebral Arteries/pathology , Cicatrix/metabolism , Cystatin C/genetics , DNA-Binding Proteins/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Male , Microfilament Proteins , Middle Aged , Neuroimmunomodulation/physiology , Young Adult
20.
EMBO Mol Med ; 7(7): 895-903, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25978969

ABSTRACT

Abnormalities in brains of Alzheimer's disease (AD) patients are thought to start long before the first clinical symptoms emerge. The identification of affected individuals at this 'preclinical AD' stage relies on biomarkers such as decreased levels of the amyloid-ß peptide (Aß) in the cerebrospinal fluid (CSF) and positive amyloid positron emission tomography scans. However, there is little information on the longitudinal dynamics of CSF biomarkers, especially in the earliest disease stages when therapeutic interventions are likely most effective. To this end, we have studied CSF Aß changes in three Aß precursor protein transgenic mouse models, focusing our analysis on the initial Aß deposition, which differs significantly among the models studied. Remarkably, while we confirmed the CSF Aß decrease during the extended course of brain Aß deposition, a 20-30% increase in CSF Aß40 and Aß42 was found around the time of the first Aß plaque appearance in all models. The biphasic nature of this observed biomarker changes stresses the need for longitudinal biomarker studies in the clinical setting and the search for new 'preclinical AD' biomarkers at even earlier disease stages, by using both mice and human samples. Ultimately, our findings may open new perspectives in identifying subjects at risk for AD significantly earlier, and in improving the stratification of patients for preventive treatment strategies.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Cerebrospinal Fluid/chemistry , Animals , Brain/pathology , Disease Models, Animal , Early Diagnosis , Humans , Longitudinal Studies , Mice , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...