Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 33
1.
J Neuroinflammation ; 15(1): 170, 2018 May 29.
Article En | MEDLINE | ID: mdl-29843759

BACKGROUND: While most patients with Alzheimer's disease (AD) present with memory complaints, 30% of patients with early disease onset present with non-amnestic symptoms. This atypical presentation is thought to be caused by a different spreading of neurofibrillary tangles (NFT) than originally proposed by Braak and Braak. Recent studies suggest a prominent role for neuroinflammation in the spreading of tau pathology. METHODS: We aimed to explore whether an atypical spreading of pathology in AD is associated with an atypical distribution of neuroinflammation. Typical and atypical AD cases were selected based on both NFT distribution and amnestic or non-amnestic clinical presentation. Immunohistochemistry was performed on the temporal pole and superior parietal lobe of 10 typical and 9 atypical AD cases. The presence of amyloid-beta (N-terminal; IC16), pTau (AT8), reactive astrocytes (GFAP), microglia (Iba1, CD68, and HLA-DP/DQ/DR), and complement factors (C1q, C3d, C4b, and C5b-9) was quantified by image analysis. Differences in lobar distribution patterns of immunoreactivity were statistically assessed using a linear mixed model. RESULTS: We found a temporal dominant distribution for amyloid-beta, GFAP, and Iba1 in both typical and atypical AD. Distribution of pTau, CD68, HLA-DP/DQ/DR, C3d, and C4b differed between AD variants. Typical AD cases showed a temporal dominant distribution of these markers, whereas atypical AD cases showed a parietal dominant distribution. Interestingly, when quantifying for the number of amyloid-beta plaques instead of stained surface area, atypical AD cases differed in distribution pattern from typical AD cases. Remarkably, plaque morphology and localization of neuroinflammation within the plaques was different between the two phenotypes. CONCLUSIONS: Our data show a different localization of neuroinflammatory markers and amyloid-beta plaques between AD phenotypes. In addition, these markers reflect the atypical distribution of tau pathology in atypical AD, suggesting that neuroinflammation might be a crucial link between amyloid-beta deposits, tau pathology, and clinical symptoms.


Alzheimer Disease/pathology , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Parietal Lobe/metabolism , Aged , Aged, 80 and over , Amyloid beta-Peptides/metabolism , Autopsy , Calcium-Binding Proteins , Complement System Proteins/metabolism , Female , Humans , Male , Microfilament Proteins , Middle Aged , Neurofibrillary Tangles/metabolism , tau Proteins/metabolism
2.
Alzheimers Res Ther ; 9(1): 28, 2017 Apr 17.
Article En | MEDLINE | ID: mdl-28412965

BACKGROUND: Progressive supranuclear palsy (PSP) with a frontal presentation, characterized by cognitive deficits and behavioral changes, has been recognized as an early clinical picture, distinct from the classical so-called Richardson and parkinsonism presentations. The midcingulate cortex is associated with executive and attention tasks and has consistently been found to be impaired in imaging studies of patients with PSP. The aim of the present study was to determine alterations in neurotransmission underlying the pathophysiology of PSP, as well as their significance for clinically identifiable PSP subgroups. METHODS: In vitro receptor autoradiography was used to quantify densities of 20 different receptors in the caudate nucleus and midcingulate area 24' of patients with PSP (n = 16) and age- and sex-matched control subjects (n = 14). RESULTS: Densities of γ-aminobutyric acid type B, peripheral benzodiazepine, serotonin receptor type 2, and N-methyl-D-aspartate receptors were significantly higher in area 24' of patients with PSP, where tau impairment was stronger than in the caudate nucleus. Kainate and nicotinic cholinergic receptor densities were significantly lower, and adenosine receptor type 1 (A1) receptors significantly higher, in the caudate nucleus of patients with PSP. Receptor fingerprints also segregated PSP subgroups when clinical parameters such as occurrence of frontal presentation and tau pathology severity were taken into consideration. CONCLUSIONS: We demonstrate, for the first time to our knowledge, that kainate and A1 receptors are altered in PSP and that clinically identifiable PSP subgroups differ at the neurochemical level. Numerous receptors were altered in the midcingulate cortex, further suggesting that it may prove to be a key region in PSP. Finally, we add to the evidence that nondopaminergic systems play a role in the pathophysiology of PSP, thus highlighting potential novel treatment strategies.


Caudate Nucleus/metabolism , Gyrus Cinguli/metabolism , Receptors, Neurotransmitter/metabolism , Supranuclear Palsy, Progressive/metabolism , Aged , Aged, 80 and over , Autoradiography , Caudate Nucleus/pathology , Cohort Studies , Female , Gyrus Cinguli/pathology , Humans , Male , Middle Aged , Severity of Illness Index , Supranuclear Palsy, Progressive/pathology , Synaptic Transmission/physiology , tau Proteins/metabolism
3.
Brain ; 137(Pt 5): 1361-73, 2014 May.
Article En | MEDLINE | ID: mdl-24722252

Pathological accumulation of intermediate filaments can be observed in neurodegenerative disorders, such as Alzheimer's disease, frontotemporal dementia and Parkinson's disease, and is also characteristic of neuronal intermediate filament inclusion disease. Intermediate filaments type IV include three neurofilament proteins (light, medium and heavy molecular weight neurofilament subunits) and α-internexin. The phosphorylation of intermediate filament proteins contributes to axonal growth, and is regulated by protein kinase A. Here we describe a family with a novel late-onset neurodegenerative disorder presenting with dementia and/or parkinsonism in 12 affected individuals. The disorder is characterized by a unique neuropathological phenotype displaying abundant neuronal inclusions by haematoxylin and eosin staining throughout the brain with immunoreactivity for intermediate filaments. Combining linkage analysis, exome sequencing and proteomics analysis, we identified a heterozygous c.149T>G (p.Leu50Arg) missense mutation in the gene encoding the protein kinase A type I-beta regulatory subunit (PRKAR1B). The pathogenicity of the mutation is supported by segregation in the family, absence in variant databases, and the specific accumulation of PRKAR1B in the inclusions in our cases associated with a specific biochemical pattern of PRKAR1B. Screening of PRKAR1B in 138 patients with Parkinson's disease and 56 patients with frontotemporal dementia did not identify additional novel pathogenic mutations. Our findings link a pathogenic PRKAR1B mutation to a novel hereditary neurodegenerative disorder and suggest an altered protein kinase A function through a reduced binding of the regulatory subunit to the A-kinase anchoring protein and the catalytic subunit of protein kinase A, which might result in subcellular dislocalization of the catalytic subunit and hyperphosphorylation of intermediate filaments.


Cyclic AMP-Dependent Protein Kinase RIbeta Subunit/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Polymorphism, Single Nucleotide/genetics , Aged , Amyloid beta-Peptides/metabolism , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/chemistry , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , DNA-Binding Proteins/metabolism , Electron Microscope Tomography , Family Health , Female , Frontal Lobe/metabolism , Frontal Lobe/pathology , Frontal Lobe/ultrastructure , Genetic Association Studies , Humans , Male , Middle Aged , Models, Molecular , Nerve Tissue Proteins/metabolism , alpha-Synuclein/metabolism , tau Proteins/metabolism
4.
J Alzheimers Dis ; 33(3): 715-21, 2013.
Article En | MEDLINE | ID: mdl-23001706

Neuropsychiatric symptoms (NPSs) have a large impact on the quality of life of patients with dementia. A few studies have compared neuropsychiatric disturbances between dementia subtypes, but the results were conflicting. In the present study, we investigated whether the prevalence of NPSs differs between Alzheimer's disease (AD) and vascular dementia (VaD). The merit of our study is that we used clinical as well as histopathological information to differentiate between dementia subtypes. This retrospective descriptive study comprised 80 brains obtained from donors to the Netherlands Brain Bank between 1984 and 2010. These donors were diagnosed postmortem with AD (n = 40) or VaD (n = 40). We assessed the presence of NPSs by reviewing the information found in the patients' medical files. The most prevalent symptom in the sample as a whole was agitation (45 cases, 57.0%), followed by depression (33, 41.2%) and anxiety (28, 35.4%). Our study tried to contribute to the discussion by including, for the first time in the literature, a sample of AD and VaD patients with neuropathologically confirmed diagnoses. Since no significant differences were found between AD and VaD patients, we suggest that the prevalence of NPSs cannot be predicted from the diagnosis of AD or VaD.


Alzheimer Disease/complications , Behavioral Symptoms/etiology , Dementia, Vascular/complications , Mental Disorders/etiology , Aged , Aged, 80 and over , Behavioral Symptoms/diagnosis , Female , Humans , Logistic Models , Male , Mental Disorders/diagnosis , Neuropsychological Tests , Psychiatric Status Rating Scales , Retrospective Studies , Severity of Illness Index
5.
J Neuropathol Exp Neurol ; 70(8): 698-702, 2011 Aug.
Article En | MEDLINE | ID: mdl-21760536

A rare case of Gerstmann-Sträussler-Scheinker disease in a 36-year-old Dutch man is reported. The clinical phenotype was characterized by slowly progressive cognitive decline, later followed by ataxia and parkinsonism. Neuropathologic findings consisted of numerous amyloid plaques in the cerebellum, which showed positive staining for the abnormal prion protein (PrP(Sc)). In addition, there were tau accumulations around numerous amyloid deposits in the cerebral cortex, striatum, hippocampal formation, and midbrain. There was no spongiform degeneration. Western blot analysis showed the co-occurrence of 2 distinct abnormal prion protein species comprising an unglycosylated, protease-resistant fragment of approximately 8 kd, which was found to be truncated at both N- and C-terminal ends by epitope mapping, and a detergent-insoluble but protease-sensitive form of full-length PrP(Sc). Sequence analysis disclosed a mutation at codon 131 of the prion protein gene (PRNP), resulting in a valine-for-glycine substitution (G131V). The patient was heterozygous at the polymorphic codon 129 and carried the mutation on the methionine allele. To our knowledge, this is the second family worldwide in which this mutation has been identified. Gerstmann-Sträussler-Scheinker disease should be considered in patients with a clinical diagnosis of familial frontotemporal dementia.


Genetic Predisposition to Disease , Gerstmann-Straussler-Scheinker Disease/genetics , Glycine/genetics , Polymorphism, Genetic/genetics , Prions/genetics , Valine/genetics , Adult , Brain/metabolism , Brain/pathology , Genome-Wide Association Study , Gerstmann-Straussler-Scheinker Disease/pathology , Gerstmann-Straussler-Scheinker Disease/physiopathology , Humans , Male , Netherlands , Prions/metabolism
6.
J Neurol Neurosurg Psychiatry ; 81(4): 441-5, 2010 Apr.
Article En | MEDLINE | ID: mdl-20360166

OBJECTIVE: To compare survival and to identify prognostic predictors for progressive supranuclear palsy and frontotemporal dementia. BACKGROUND: Progressive supranuclear palsy (PSP) and frontotemporal dementia (FTD) are related disorders. Homozygosity for H1 haplotype is associated with PSP, whereas several MAPT mutations have been identified in FTLD-tau. Survival duration probably reflects underlying pathophysiology or disease. METHODS: Patients with PSP and FTD were recruited by nationwide referral. Survival of 354 FTD patients was compared with that of 197 PSP patients. Cox regression analysis was performed to identify prognostic predictors. FTLD-tau was defined as Pick disease and FTDP-17 with MAPT mutations. Semiquantitative evaluation of tau-positive pathology was performed on all pathologically proven cases. RESULTS: The median survival of PSP patients (8.0 years; 95% CI 7.3 to 8.7) was significantly shorter than that of FTD patients (9.9 years; 95% CI 9.2 to 10.6). Corrected for demographic differences, PSP patients were still significantly more at risk of dying than FTD patients. In PSP, male gender, older onset-age and higher PSP Rating Scale score were identified as independent predictors for shorter survival, whereas in FTD a positive family history and an older onset-age were associated with a poor prognosis. The difference in hazard rate was even more pronounced when comparing pathologically proven cases of PSP with FTLD-tau. CONCLUSION: Survival of PSP patients is shorter than that of FTD patients, and probably reflects a more aggressive disease process in PSP. Independent predictors of shorter survival in PSP were male gender, older onset-age and higher PSP rating scale score, whereas in FTD a positive family history and higher onset-age were predictors for worse prognosis.


Frontotemporal Dementia/mortality , Supranuclear Palsy, Progressive/mortality , Age of Onset , Aged , Brain/blood supply , Brain/pathology , Cognition Disorders/diagnosis , Cognition Disorders/epidemiology , Female , Frontotemporal Dementia/diagnosis , Frontotemporal Dementia/genetics , Genotype , Humans , Magnetic Resonance Imaging , Male , Neuropsychological Tests , Prognosis , Prospective Studies , Supranuclear Palsy, Progressive/diagnosis , Supranuclear Palsy, Progressive/genetics , Survival Rate , Time Factors , Tomography, Emission-Computed, Single-Photon , tau Proteins/genetics
7.
Acta Neuropsychiatr ; 22(1): 14-20, 2010 Feb.
Article En | MEDLINE | ID: mdl-25384952

UNLABELLED: Meynen G, Van Stralen H, Smit JH, Kamphorst W, Swaab DF, Hoogendijk WJG. Relation between neuritic plaques and depressive state in Alzheimer's disease. BACKGROUND: To investigate for the first time in a prospective study the relationship between depressive state and the neuropathological hallmarks of Alzheimer's disease, using a scale for depressive symptoms in dementia, while controlling for clinical severity of dementia. METHOD: Within the framework of a prospective longitudinal study of depression in Alzheimer's disease, patients with dementia underwent a clinical evaluation every six months during the last years of their lives, using the Cornell scale for depression in dementia to assess depressive symptoms and using the Functional Assessment Staging scale to control for clinical severity of dementia. The brains of 43 Alzheimer patients were obtained. The last clinical evaluations prior to death together with post-mortem neuropathology measures were analysed. RESULTS: We found a correlation between the Cornell scores and the sum score for the density of neuritic plaques in the entire cortex (p = 0.027), and even stronger in the temporal cortex (p = 0.012). The observed correlations were independent of sex, age of death, clinical dementia severity and duration of Alzheimer's disease. CONCLUSIONS: This study shows a positive relationship between depressive state at time of death and the presence of neuritic plaques in Alzheimer's disease, which is independent of the clinical severity of dementia.

9.
PLoS One ; 4(8): e6826, 2009 Aug 28.
Article En | MEDLINE | ID: mdl-19714246

BACKGROUND: Understanding the aetiologies of neurodegenerative diseases such as Alzheimer's disease (AD), Pick's disease (PiD), Progressive Supranuclear Palsy (PSP) and Frontotemporal dementia (FTD) is often hampered by the considerable clinical and molecular overlap between these diseases and normal ageing. The development of high throughput genomic technologies such as microarrays provide a new molecular tool to gain insight in the complexity and relationships between diseases, as they provide data on the simultaneous activity of multiple genes, gene networks and cellular pathways. METHODOLOGY/PRINCIPAL FINDINGS: We have constructed genome wide expression profiles from snap frozen post-mortem tissue from the medial temporal lobe of patients with four neurodegenerative disorders (5 AD, 5 PSP, 5 PiD and 5 FTD patients) and 5 control subjects. All patients were matched for age, gender, ApoE-epsilon and MAPT (tau) haplotype. From all groups a total of 790 probes were shown to be differently expressed when compared to control individuals. The results from these experiments were then used to investigate the correlations between clinical, pathological and molecular findings. From the 790 identified probes we extracted a gene set of 166 probes whose expression could discriminate between these disorders and normal ageing. CONCLUSIONS/SIGNIFICANCE: From genome wide expression profiles we extracted a gene set of 166 probes whose expression could discriminate between neurological disorders and normal ageing. This gene set can be further developed into an accurate microarray-based classification test. Furthermore, from this dataset we extracted a disease specific set of genes and identified two aging related transcription factors (FOXO1A and FOXO3A) as possible drug targets related to neurodegenerative disease.


Gene Expression Profiling , Neurodegenerative Diseases/genetics , RNA, Messenger/genetics , tau Proteins/physiology , Case-Control Studies , Cluster Analysis , DNA Primers , Genome-Wide Association Study , Humans , Neurodegenerative Diseases/physiopathology , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , tau Proteins/genetics
11.
Brain Pathol ; 18(4): 484-96, 2008 Oct.
Article En | MEDLINE | ID: mdl-18371174

It has been recognized that molecular classifications will form the basis for neuropathological diagnostic work in the future. Consequently, in order to reach a diagnosis of Alzheimer's disease (AD), the presence of hyperphosphorylated tau (HP-tau) and beta-amyloid protein in brain tissue must be unequivocal. In addition, the stepwise progression of pathology needs to be assessed. This paper deals exclusively with the regional assessment of AD-related HP-tau pathology. The objective was to provide straightforward instructions to aid in the assessment of AD-related immunohistochemically (IHC) detected HP-tau pathology and to test the concordance of assessments made by 25 independent evaluators. The assessment of progression in 7-microm-thick sections was based on assessment of IHC labeled HP-tau immunoreactive neuropil threads (NTs). Our results indicate that good agreement can be reached when the lesions are substantial, i.e., the lesions have reached isocortical structures (stage V-VI absolute agreement 91%), whereas when only mild subtle lesions were present the agreement was poorer (I-II absolute agreement 50%). Thus, in a research setting when the extent of lesions is mild, it is strongly recommended that the assessment of lesions should be carried out by at least two independent observers.


Alzheimer Disease/pathology , Brain/pathology , Neurofibrillary Tangles/pathology , Neurons/pathology , tau Proteins/metabolism , Adult , Age Factors , Aged , Aged, 80 and over , Aging/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Brain/metabolism , Brain/physiopathology , Disease Progression , Female , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Immunohistochemistry/methods , Male , Middle Aged , Neocortex/metabolism , Neocortex/pathology , Neocortex/physiopathology , Neurofibrillary Tangles/metabolism , Neurons/metabolism , Observer Variation , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Staining and Labeling/methods , tau Proteins/analysis
12.
J Neuropathol Exp Neurol ; 67(2): 125-43, 2008 Feb.
Article En | MEDLINE | ID: mdl-18219257

To determine the reliability of assessment of alpha-synuclein-immunoreactive (alphaS-IR) structures by neuropathologists, 28 evaluators from 17 centers of BrainNet Europe examined current methods and reproducibility of alphaS-IR evaluation using a tissue microarray (TMA) technique. Tissue microarray blocks were constructed of samples from the participating centers that contained alphaS-IR structures. Slides from these blocks were stained in each center and assessed for neuronal perikaryal inclusions, neurites, and glial cytoplasmic inclusions. The study was performed in 2 phases. First, the TMA slides were stained with the antibody of the center's choice. In this phase, 59% of the sections were of good or acceptable quality, and 4 of 9 antibodies used performed consistently. Differences in interpretation and categorization of alphaS-IR structures, however, led to differing results between the laboratories. Prior to the second phase, the neuropathologists participated in a training session on the evaluation of alphaS-IR structures. Based on the results of the first phase, selected antibodies using designated antigen retrieval methods were then applied to TMA slides in the second phase. When the designated methods of both staining and evaluation were applied, all 26 subsequently stained TMA sections evaluated were of good/acceptable quality, and a high level of concordance in the assessment of the presence or absence of specific alphaS-IR structures was achieved. A semiquantitative assessment of alphaS-IR neuronal perikaryal inclusions yielded agreements ranging from 49% to 82%, with best concordance in cortical core samples. These results suggest that rigorous methodology and dichotomized assessment (i.e. determining the presence or absence of alphaS-IR) should be applied, and that semiquantitative assessment can be recommended only for the cortical samples. Moreover, the study demonstrates that there are limitations in the scoring of alphaS-IR structures.


Brain Diseases/pathology , Brain/metabolism , Brain/pathology , Database Management Systems , alpha-Synuclein/metabolism , Adult , Aged , Aged, 80 and over , Database Management Systems/statistics & numerical data , Europe , Female , Humans , Immunohistochemistry , Male , Microarray Analysis/methods , Middle Aged , Neuroglia/metabolism , Neurons/metabolism , Statistics, Nonparametric
13.
J Neuroimmunol ; 190(1-2): 157-64, 2007 Oct.
Article En | MEDLINE | ID: mdl-17884183

Chemokines mediate selective recruitment of leukocyte subsets into the CNS during inflammatory episodes. We hypothesised that functional polymorphisms in CCR5 and CCL5 influence perivascular leukocyte infiltration, inflammation, axonal loss, and remyelination, and disease course. Therefore, we determined genotypes at four possibly functional polymorphisms in CCR5 and CCL5 for 637 patients and 92 brain donors with multiple sclerosis (MS). For a subset of 192 patients, MRI data were available. We found that low-producer allele CCL5-403*G was associated with reduced risk of severe axonal loss, whereas high-producer allele CCL5-403*A was associated with a worse clinical disease course measured by the MS Functional Composite Score and MS Severity Score. Low-producer allele CCR5+303*G was associated with reduced T2 hyperintense and T1 hypointense lesion volumes on MRI, and high-producer allele CCR5+303*A with early age at onset. Furthermore, low-producer allele CCR5Delta32 was associated with reduced T2 lesion volume, lower black hole ratio on MRI, and with a higher percentage of lesions with signs of remyelination, histopathologically. In summary, our multifaceted study supports the notion that polymorphisms in CCL5 and CCR5 modify the course of MS.


Central Nervous System/pathology , Chemokine CCL5/genetics , Genetic Predisposition to Disease/genetics , Multiple Sclerosis/genetics , Receptors, CCR5/genetics , Adult , Aged , Central Nervous System/diagnostic imaging , Central Nervous System/physiopathology , Chemokine CCL5/immunology , Chemotaxis, Leukocyte/genetics , DNA Mutational Analysis , Disease Progression , Female , Gene Frequency/genetics , Genetic Markers/genetics , Genetic Testing , Genotype , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , Myelin Sheath/immunology , Myelin Sheath/pathology , Predictive Value of Tests , Prognosis , Radiography , Receptors, CCR5/immunology , Sensitivity and Specificity , Wallerian Degeneration/genetics , Wallerian Degeneration/immunology , Wallerian Degeneration/physiopathology
14.
Acta Neuropathol ; 114(4): 403-10, 2007 Oct.
Article En | MEDLINE | ID: mdl-17628813

Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a progressive cerebral white matter disease with onset in childhood, caused by mutations in the MLC1 gene. MLC1 is a protein with unknown function that is mainly expressed in the brain in astrocytic endfeet at the blood-brain and cerebrospinal fluid-brain barriers. It shares its localization at astrocytic endfeet with the dystrophin-associated glycoprotein complex (DGC). The objective of the present study was to investigate the possible association of MLC1 with the DGC. To test this hypothesis, (co)-localization of DGC-proteins and MLC1 was analyzed by immunohistochemical stainings in gliotic brain tissue from a patient with multiple sclerosis, in glioblastoma tissue and in brain tissue from an MLC patient. In control tissue, a direct protein interaction was tested by immunoprecipitation. Results revealed that MLC1 is co-localized with DGC-proteins in gliotic brain tissue. We demonstrated that both MLC1 and aquaporin-4, a member of the DGC, were redistributed in glioblastoma cells. In MLC brain tissue, we showed absence of MLC1 and altered expression of several DGC-proteins. We demonstrated a direct protein interaction between MLC1 and Kir4.1. From these results we conclude that MLC1 is associated with the DGC at astrocytic endfeet.


Astrocytes/metabolism , Brain/metabolism , Central Nervous System Cysts/metabolism , Dystrophin-Associated Protein Complex/metabolism , Heredodegenerative Disorders, Nervous System/metabolism , Membrane Proteins/metabolism , Aquaporin 4/metabolism , Astrocytes/pathology , Blotting, Western , Brain/pathology , Brain Neoplasms/metabolism , Central Nervous System Cysts/pathology , Glioma/metabolism , Glycoproteins/metabolism , Heredodegenerative Disorders, Nervous System/pathology , Humans , Immunohistochemistry , Immunoprecipitation , Multiple Sclerosis/metabolism , Potassium Channels, Inwardly Rectifying/metabolism
15.
Brain ; 130(Pt 5): 1375-85, 2007 May.
Article En | MEDLINE | ID: mdl-17360763

Frontotemporal dementia is accompanied by motor neuron disease (FTD + MND) in approximately 10% of cases. There is accumulating evidence for a clinicopathological overlap between FTD and MND based on observations of familial aggregation and neuropathological findings of ubiquitin-positive neuronal cytoplasmatic inclusions (NCI) in lower motor neurons, hippocampus and neocortex in both conditions. Several familial forms exist with different genetic loci and defects. We investigated the familial aggregation and clinical presentation of FTD + MND cases in a large cohort of 368 FTD patients in The Netherlands. Immunohistochemistry of available brain tissue of deceased patients was investigated using a panel of antibodies including ubiquitin, p62 and TAR DNA-binding protein of 43 kDa antibodies. A total of eight patients coming from six families had a family history positive for FTD + MND (mean age at onset 53.2 +/- 8.4 years). Five patients presented with behavioural changes and cognitive changes followed by motor neuron disease, whereas symptoms of motor neuron disease were the presenting features in the remaining three patients. Other affected relatives in these families showed dementia/FTD, MND or FTD + MND reflecting the clinical interfamilial variation. No mutations were identified in any of the candidate genes, including Superoxide Dismutase 1, dynactin, angiogenin, Microtubule-Associated Protein Tau, valosin-containing protein and progranulin. Available brain tissue of five patients with familial FTD + MND showed NCI in hippocampus, neocortex and spinal cord in all, and neuronal intranuclear inclusions (NII) in two brains. TDP-43 antibody showed robust staining of neuronal inclusions similar in distribution and morphology to NCI and NII. Additionally, TDP-43 antibody also stained ubiquitin-negative glial inclusions in the basal striatum of one case. In conclusion, there exists considerable clinical variation within families with FTD + MND, which may be determined by other genetic or environmental factors. NII are also found in some cases of familial FTD + MND without Progranulin mutations. The observation of glial TDP-43 positive inclusions in one brain is very interesting, although their pathophysiological significance is yet unknown.


Brain Chemistry , DNA-Binding Proteins/analysis , Motor Neuron Disease/complications , Pick Disease of the Brain/complications , Adaptor Proteins, Signal Transducing/analysis , Adult , Cohort Studies , Female , Genotype , Hippocampus/chemistry , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Male , Middle Aged , Motor Neuron Disease/genetics , Motor Neuron Disease/metabolism , Motor Neurons/chemistry , Mutation , Neocortex/chemistry , Pedigree , Pick Disease of the Brain/genetics , Pick Disease of the Brain/metabolism , Progranulins , RNA-Binding Proteins , Sequestosome-1 Protein , Ubiquitin/analysis
16.
J Neuropathol Exp Neurol ; 66(1): 17-25, 2007 Jan.
Article En | MEDLINE | ID: mdl-17204933

Tau mutations in frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) are associated with changes in alternative splicing of exon 10. The DeltaK280 mutation in exon 10 is exceptional because in vitro observations suggest a dramatic effect on microtubule binding, enhanced self-aggregation, as well as a decrease of the 4R/3R ratio by the ablation of an exon splicing enhancer element. Using immunohistochemistry, Western blotting, and electron microscopy on brain material with the DeltaK280 mutation, we investigated which of these effects is most dominant in vivo. The brain showed abundant Pick bodies in several brain regions, which stained positive with 3-repeat-specific but not with 4-repeat-specific tau antibodies. Western blots of sarkosyl-insoluble tau showed exclusively three repeat (3R0N and 3R1N) tau in most regions, although some 4R1N could be detected in the frontal cortex. In addition, the sarkosyl-soluble tau fraction showed a significantly higher amount of 3-repeat tau. Because quantitative analysis of 4R and 3R mRNA transcripts showed a 4R/3R ratio of only 0.3, association between increased transcription and protein expression was observed. These observations confirm the postulated hypothesis that the DeltaK280 mutation abolishes a splice enhancer element, which overrules the decreased microtubule binding and enhanced self-aggregation.


Dementia/genetics , Exons , Lysine/genetics , Mutation , tau Proteins/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Blotting, Western/methods , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Dementia/pathology , Female , Humans , Immunohistochemistry/methods , Microscopy, Electron, Transmission/methods , Middle Aged , Molecular Weight , Neurons/metabolism , Neurons/pathology , Serine/metabolism , Trinucleotide Repeat Expansion/genetics
17.
Acta Neuropathol ; 113(4): 389-402, 2007 Apr.
Article En | MEDLINE | ID: mdl-17237937

Amyloid beta (Abeta) immunoreactivity in neurons was examined in brains of 32 control subjects, 31 people with Down syndrome, and 36 patients with sporadic Alzheimer's disease to determine if intraneuronal Abeta immunoreactivity is an early manifestation of Alzheimer-type pathology leading to fibrillar plaque formation and/or neurofibrillary degeneration. The appearance of Abeta immunoreactivity in neurons in infants and stable neuron-type specific Abeta immunoreactivity in a majority of brain structures during late childhood, adulthood, and normal aging does not support this hypothesis. The absence or detection of only traces of reaction with antibodies against 4-13 aa and 8-17 aa of Abeta in neurons indicated that intraneuronal Abeta was mainly a product of alpha- and gamma-secretases (Abeta(17-40/42)). The presence of N-terminally truncated Abeta(17-40) and Abeta(17-42) in the control brains was confirmed by Western blotting and the identity of Abeta(17-40) was confirmed by mass spectrometry. The prevalence of products of alpha- and gamma -secretases in neurons and beta- and gamma-secretases in plaques argues against major contribution of Abeta-immunopositive material detected in neuronal soma to amyloid deposit in plaques. The strongest intraneuronal Abeta(17-42) immunoreactivity was observed in structures with low susceptibility to fibrillar Abeta deposition, neurofibrillary degeneration, and neuronal loss compared to areas more vulnerable to Alzheimer-type pathology. These observations indicate that the intraneuronal Abeta immunoreactivity detected in this study is not a predictor of brain amyloidosis or neurofibrillary degeneration. The constant level of Abeta immunoreactivity in structures free from neuronal pathology during essentially the entire life span suggests that intraneuronal amino-terminally truncated Abeta represents a product of normal neuronal metabolism.


Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/pathology , Down Syndrome/metabolism , Intracellular Fluid/metabolism , Neurofibrillary Tangles/pathology , Neurons/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Case-Control Studies , Child, Preschool , Down Syndrome/pathology , Female , Humans , Infant , Male , Middle Aged , Predictive Value of Tests
18.
J Neuropathol Exp Neurol ; 65(8): 740-57, 2006 Aug.
Article En | MEDLINE | ID: mdl-16896308

This interlaboratory study evaluated the reproducibility of the assessments of neuritic plaques and neurofibrillary tangles (NFTs)--the hallmark lesions of Alzheimer disease--and compared the staining between the BrainNet Europe centers. To reduce the topography-related inconsistencies in assessments, we used a 2-mm tissue microarray (TMA) technique. The TMA block included 42 core samples taken from 21 paraffin blocks. The assessments were done on Bielschowsky and Gallyas silver stains using an immunohistochemical (IHC) method with antibodies directed to beta-amyloid (IHC/Abeta) and hyperphosphorylated tau (IHC/HPtau). The staining quality and the assessments differed between the participants, being most diverse with Bielschowsky (good/acceptable stain in 53% of centers) followed by Gallyas (good/acceptable stain in 57%) and IHC/Abeta (good/acceptable stain in 71%). The most uniform staining quality and assessment was obtained with the IHC/HPtau method (good/acceptable stain in 94% of centers). The neuropathologic diagnostic protocol (Consortium to Establish a Registry for Alzheimer Disease, Braak and Braak, and the National Institute of Aging and Reagan [NIA-Reagan] Institute) that was used significantly influenced the agreement, being highest with NIA-Reagan (54%) recommendations. This agreement was improved by visualization of NFTs using the IHC/HPtau method. Therefore, the IHC/HPtau methodology to visualize NFTs and neuropil threads should be considered as a method of choice in a future diagnostic protocol for Alzheimer disease.


Alzheimer Disease/diagnosis , Cerebral Cortex/pathology , Neurofibrillary Tangles/pathology , Plaque, Amyloid/pathology , Staining and Labeling/standards , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/metabolism , Biopsy/methods , Biopsy/standards , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Europe , Female , Humans , Immunohistochemistry/methods , Immunohistochemistry/standards , International Agencies/standards , International Agencies/statistics & numerical data , Male , Middle Aged , Neurofibrillary Tangles/metabolism , Pathology/methods , Pathology/standards , Plaque, Amyloid/metabolism , Registries/standards , Registries/statistics & numerical data , Silver Staining/methods , Silver Staining/standards , Staining and Labeling/methods , Tissue Banks/standards , Tissue Banks/statistics & numerical data , tau Proteins/analysis , tau Proteins/metabolism
19.
J Neuropathol Exp Neurol ; 64(5): 412-9, 2005 May.
Article En | MEDLINE | ID: mdl-15892299

Megaloencephalic leukoencephalopathy with subcortical cysts (MLC) is a progressive cerebral white matter disease in children caused by mutations in the MLC1 gene. This disease is histopathologically characterized by myelin splitting and intramyelinic vacuole formation. MLC1 encodes a novel protein, MLC1, which is mainly expressed in the brain and leukocytes. The function is unknown, although a transport function has been suggested. In this article, we provide experimental data addressing the membrane topology and cellular localization of MLC1. We show that MLC1 contains an even number of transmembrane domains, supporting the possible transport function of MLC1. We demonstrate that MLC1 is specifically expressed in distal astroglial processes in perivascular, subependymal, and subpial regions. This localization suggests a role for MLC1 in a transport process across the blood-brain and brain-cerebrospinal fluid barriers. Astrocyte functions have long been debated. It is becoming increasingly clear that these cells are of fundamental importance in maintaining the structural and functional integrity of neural tissue. Elucidation of the function of MLC1 will contribute to a better understanding of not only the pathophysiology of the disease, but also the role of astrocytes in normal neural tissue.


Astrocytes/metabolism , Gene Expression Regulation/physiology , Membrane Proteins/metabolism , Amino Acid Sequence , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Blotting, Western/methods , Brain/cytology , Brain/metabolism , Cell Line, Transformed , Cells, Cultured , Chickens , Cloning, Molecular/methods , Flow Cytometry/methods , Fluorescent Antibody Technique/methods , Glial Fibrillary Acidic Protein/metabolism , Humans , Intracellular Space/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Transgenic , RNA, Messenger/biosynthesis , Rats , Reverse Transcriptase Polymerase Chain Reaction/methods , Sequence Alignment , Takifugu , Transfection/methods
20.
J Neuropathol Exp Neurol ; 63(12): 1243-54, 2004 Dec.
Article En | MEDLINE | ID: mdl-15624761

Previously, alterations in neuronal metabolism were found in a number of brain areas of Alzheimer disease (AD) patients. In the present study we aimed at determining for the first time whether metabolic changes would also occur in vascular dementia (VD) patients in the supraoptic (SON), infundibular (INF), tuberomamillary (TMN), medial mamillary nuclei, vertical limb of the diagonal band of Broca (VDB), and nucleus basalis of Meynert. The Golgi complex (GC) size, cell size, and vasopressin mRNA levels (in the SON) were used as measures of neuronal metabolic activity in postmortem material. The GC immunoreactivity was clearly diminished in the SON, INF and TMN and was increased in the VDB of VD cases. Interestingly, in the SON and TMN, a decrease in the GC size was more pronounced in male than in female VD patients in accordance with the higher prevalence of VD in men. In 7 of 11 VD cases, vasopressin mRNA levels were significantly reduced which may contribute to urinary incontinence, one of the most common clinical symptoms in VD, and to the lower blood pressure values that are often registered at the later stages of the VD. Since the human TMN is the sole source of cerebral histamine, our data suggest deficient histaminergic transmission in the brain in VD. Diminished neuronal metabolism in the SON and INF was not observed in AD in this and previous studies, whereas the changes in the VDB and TMN are similar in VD and AD. In the present study we thus found decreased metabolic activity in several hypothalamic nuclei in VD indicating diminished production of certain hormones and neurotransmitters.


Dementia, Vascular/metabolism , Golgi Apparatus/metabolism , Hypothalamus/metabolism , Prosencephalon/metabolism , Vasopressins/metabolism , Aged , Aged, 80 and over , Cadaver , Cell Size , Dementia, Vascular/pathology , Female , Golgi Apparatus/pathology , Humans , Hypothalamus/pathology , In Situ Hybridization , Male , Middle Aged , Prosencephalon/pathology , RNA, Messenger/metabolism , Tissue Distribution , Vasopressins/genetics
...