Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 38
1.
Trials ; 24(1): 378, 2023 Jun 05.
Article En | MEDLINE | ID: mdl-37277877

BACKGROUND: Cerebral amyloid angiopathy (CAA) is a disease caused by the accumulation of the amyloid-beta protein and is a major cause of intracerebral hemorrhage (ICH) and vascular dementia in the elderly. The presence of the amyloid-beta protein in the vessel wall may induce a chronic state of cerebral inflammation by activating astrocytes, microglia, and pro-inflammatory substances. Minocycline, an antibiotic of the tetracycline family, is known to modulate inflammation, gelatinase activity, and angiogenesis. These processes are suggested to be key mechanisms in CAA pathology. Our aim is to show the target engagement of minocycline and investigate in a double-blind placebo-controlled randomized clinical trial whether treatment with minocycline for 3 months can decrease markers of neuroinflammation and of the gelatinase pathway in cerebrospinal fluid (CSF) in CAA patients. METHODS: The BATMAN study population consists of 60 persons: 30 persons with hereditary Dutch type CAA (D-CAA) and 30 persons with sporadic CAA. They will be randomized for either placebo or minocycline (15 sporadic CAA/15 D-CAA minocycline, 15 sporadic CAA/15 D-CAA placebo). At t = 0 and t = 3 months, we will collect CSF and blood samples, perform a 7-T MRI, and collect demographic characteristics. DISCUSSION: The results of this proof-of-principle study will be used to assess the potential of target engagement of minocycline for CAA. Therefore, our primary outcome measures are markers of neuroinflammation (IL-6, MCP-1, and IBA-1) and of the gelatinase pathway (MMP2/9 and VEGF) in CSF. Secondly, we will look at the progression of hemorrhagic markers on 7-T MRI before and after treatment and investigate serum biomarkers. TRIAL REGISTRATION: ClinicalTrials.gov NCT05680389. Registered on January 11, 2023.


Cerebral Amyloid Angiopathy, Familial , Cerebral Amyloid Angiopathy , Aged , Humans , Amyloid beta-Peptides , Anti-Bacterial Agents/pharmacology , Cerebral Amyloid Angiopathy/diagnostic imaging , Cerebral Amyloid Angiopathy/drug therapy , Cerebral Amyloid Angiopathy/complications , Cerebral Amyloid Angiopathy, Familial/complications , Cerebral Amyloid Angiopathy, Familial/pathology , Cerebral Hemorrhage/etiology , Gelatinases , Inflammation , Minocycline , Neuroinflammatory Diseases , Randomized Controlled Trials as Topic
2.
Int J Mol Sci ; 22(6)2021 Mar 13.
Article En | MEDLINE | ID: mdl-33805778

Plasma amyloid-beta (Aß) has long been investigated as a blood biomarker candidate for Cerebral Amyloid Angiopathy (CAA), however previous findings have been inconsistent which could be attributed to the use of less sensitive assays. This study investigates plasma Aß alterations between pre-symptomatic Dutch-type hereditary CAA (D-CAA) mutation-carriers (MC) and non-carriers (NC) using two Aß measurement platforms. Seventeen pre-symptomatic members of a D-CAA pedigree were assembled and followed up 3-4 years later (NC = 8; MC = 9). Plasma Aß1-40 and Aß1-42 were cross-sectionally and longitudinally analysed at baseline (T1) and follow-up (T2) and were found to be lower in MCs compared to NCs, cross-sectionally after adjusting for covariates, at both T1(Aß1-40: p = 0.001; Aß1-42: p = 0.0004) and T2 (Aß1-40: p = 0.001; Aß1-42: p = 0.016) employing the Single Molecule Array (Simoa) platform, however no significant differences were observed using the xMAP platform. Further, pairwise longitudinal analyses of plasma Aß1-40 revealed decreased levels in MCs using data from the Simoa platform (p = 0.041) and pairwise longitudinal analyses of plasma Aß1-42 revealed decreased levels in MCs using data from the xMAP platform (p = 0.041). Findings from the Simoa platform suggest that plasma Aß may add value to a panel of biomarkers for the diagnosis of pre-symptomatic CAA, however, further validation studies in larger sample sets are required.


Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Cerebral Amyloid Angiopathy, Familial/genetics , Peptide Fragments/genetics , Adult , Amyloid beta-Peptides/blood , Amyloid beta-Protein Precursor/blood , Asymptomatic Diseases , Biomarkers/blood , Cerebral Amyloid Angiopathy, Familial/blood , Cerebral Amyloid Angiopathy, Familial/diagnosis , Cerebral Amyloid Angiopathy, Familial/pathology , Disease Progression , Female , Gene Expression , Genes, Dominant , Heterozygote , Humans , Longitudinal Studies , Male , Middle Aged , Mutation , Neuropsychological Tests , Pedigree , Peptide Fragments/blood
3.
Proc Natl Acad Sci U S A ; 117(25): 14482-14492, 2020 06 23.
Article En | MEDLINE | ID: mdl-32518112

Cerebral amyloid angiopathy (CAA), where beta-amyloid (Aß) deposits around cerebral blood vessels, is a major contributor of vascular dysfunction in Alzheimer's disease (AD) patients. However, the molecular mechanism underlying CAA formation and CAA-induced cerebrovascular pathology is unclear. Hereditary cerebral amyloid angiopathy (HCAA) is a rare familial form of CAA in which mutations within the (Aß) peptide cause an increase in vascular deposits. Since the interaction between Aß and fibrinogen increases CAA and plays an important role in cerebrovascular damage in AD, we investigated the role of the Aß-fibrinogen interaction in HCAA pathology. Our work revealed the most common forms of HCAA-linked mutations, Dutch (E22Q) and Iowa (D23N), resulted in up to a 50-fold stronger binding affinity of Aß for fibrinogen. In addition, the stronger interaction between fibrinogen and mutant Aßs led to a dramatic perturbation of clot structure and delayed fibrinolysis. Immunofluorescence analysis of the occipital cortex showed an increase of fibrin(ogen)/Aß codeposition, as well as fibrin deposits in HCAA patients, compared to early-onset AD patients and nondemented individuals. Our results suggest the HCAA-type Dutch and Iowa mutations increase the interaction between fibrinogen and Aß, which might be central to cerebrovascular pathologies observed in HCAA.


Amyloid beta-Peptides/genetics , Brain/pathology , Cerebral Amyloid Angiopathy, Familial/pathology , Fibrin/metabolism , Fibrinogen/metabolism , Peptide Fragments/genetics , Amyloid beta-Peptides/metabolism , Cerebral Amyloid Angiopathy, Familial/genetics , Female , Fibrinogen/isolation & purification , Fibrinolysis/genetics , Humans , Male , Middle Aged , Mutation , Peptide Fragments/metabolism , Protein Binding/genetics , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
4.
Brain Pathol ; 29(6): 793-802, 2019 11.
Article En | MEDLINE | ID: mdl-30868685

In severe forms of cerebral amyloid angiopathy (CAA) pathology, vascular calcification has been observed in the cerebral cortex, both in vivo on MRI and CT, and post-mortem using histopathology. However, the pathomechanisms leading to calcification of CAA-laden arteries are unknown. Therefore, we investigated the correlation between calcification of cortical arterioles and several potential modulators of vascular calcification using immunohistochemistry in a unique collection of brain material of patients with a hereditary form of CAA, namely hereditary cerebral hemorrhage with amyloidosis-Dutch type (HCHWA-D or D-CAA). We show a topographical association of osteopontin (OPN) and TGFß signaling factor phospho-SMAD2/3 (pSMAD2/3) in calcified CAA vessel walls. OPN and pSMAD2/3 gradually accumulate in vessels prior to calcification. Moreover, we found that the vascular accumulation of Collagen 1 (Col1), OPN and pSMAD2/3 immunomarkers correlated with the CAA severity. This was independently of the vessel size, including capillaries in the most severe cases. We propose that calcification of CAA vessels in the observed HCHWA-D cases may be induced by extracellular OPN trapped in the fibrotic Col1 vessel wall, independently of the presence of vascular amyloid.


Calcinosis/pathology , Cerebral Amyloid Angiopathy/pathology , Osteopontin/metabolism , Aged , Alzheimer Disease/pathology , Amyloid , Amyloid beta-Peptides/metabolism , Amyloidosis/pathology , Arterioles/pathology , Brain/pathology , Capillaries/metabolism , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy, Familial/pathology , Cerebral Cortex/pathology , Collagen Type I/metabolism , Female , Humans , Male , Middle Aged , Osteopontin/physiology , Phosphorylation , Smad2 Protein/metabolism , Smad3 Protein/metabolism
5.
Stem Cell Res ; 34: 101359, 2019 01.
Article En | MEDLINE | ID: mdl-30611017

Hereditary Cerebral Hemorrhage with Amyloidosis-Dutch type (HCHWA-D) is an autosomal dominant hereditary disease caused by a point mutation in exon 17 of the APP gene. We generated human induced pluripotent stem cells (hiPSCs) from a symptomatic HCHWA-D patient by using non-integrating Sendai virus (SeV). The newly generated hiPSCs express all pluripotency markers, have a normal karyotype, carry the Dutch mutation, can differentiate in the three germ layers in vitro and are SeV free.


Cell Culture Techniques/methods , Cerebral Amyloid Angiopathy, Familial/pathology , Induced Pluripotent Stem Cells/pathology , Base Sequence , Cell Line , Female , Humans , Middle Aged
6.
Stroke ; 49(9): 2081-2087, 2018 09.
Article En | MEDLINE | ID: mdl-30354978

Background and Purpose- Previous studies of symptomatic and asymptomatic hereditary cerebral amyloid angiopathy (CAA) patients offered the possibility to study the radiological manifestations of CAA in the early stages of the disease. Recently, a striped cortex, observable as hypointense lines perpendicular to the pial surface on T2*-weighted 7T magnetic resonance imaging (MRI), was detected in 40% of the symptomatic hereditary CAA patients. However, the origin of these MRI contrast changes is unknown. This study aimed at defining the underlying pathology associated with the in vivo observed striped pattern. Methods- Formalin-fixed postmortem brain material including the occipital lobe of 4 hereditary cerebral hemorrhage with amyloidosis-Dutch type (HCHWA-D) cases and 6 sporadic CAA cases were selected from local neuropathology tissue collections. Depending on the availability of the material, intact hemispheres or brain slabs including the occipital lobe of these patients were screened for the presence of a striped cortex. Regions containing the striped cortex were then subjected to high-resolution 7T MRI and histopathologic examination. Results- We found 2 hereditary cerebral hemorrhage with amyloidosis-Dutch type cases and 1 sporadic CAA case with striped patterns in the occipital cortex resembling the in vivo signal. Histopathologic examination showed that the striped pattern in the cortex at 7T MRI is because of iron accumulation and calcification of penetrating arteries. The presence of both nonheme iron and calcification on penetrating arteries causes signal loss and hence the abnormal striped patterns in the cortical ribbon on T2*-weighted MRI. Conclusions- We identified iron accumulation and calcification of the vessel wall in hereditary cerebral hemorrhage with amyloidosis-Dutch type as the histopathologic correlates of the striped cortex observed on in vivo 7T MRI.


Cerebral Amyloid Angiopathy, Familial/diagnostic imaging , Iron/metabolism , Occipital Lobe/diagnostic imaging , Vascular Calcification/diagnostic imaging , Aged , Aged, 80 and over , Cerebral Amyloid Angiopathy/diagnostic imaging , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/pathology , Cerebral Amyloid Angiopathy, Familial/metabolism , Cerebral Amyloid Angiopathy, Familial/pathology , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Occipital Lobe/metabolism , Occipital Lobe/pathology , Vascular Calcification/pathology
7.
Brain Pathol ; 28(4): 495-506, 2018 07.
Article En | MEDLINE | ID: mdl-28557134

Hereditary cerebral hemorrhage with amyloidosis-Dutch type (HCHWA-D) is an early onset hereditary form of cerebral amyloid angiopathy (CAA) pathology, caused by the E22Q mutation in the amyloid ß (Aß) peptide. Transforming growth factor ß1 (TGFß1) is a key player in vascular fibrosis and in the formation of angiopathic vessels in transgenic mice. Therefore, we investigated whether the TGFß pathway is involved in HCHWA-D pathogenesis in human postmortem brain tissue from frontal and occipital lobes. Components of the TGFß pathway were analyzed with quantitative RT-PCR. TGFß1 and TGFß Receptor 2 (TGFBR2) gene expression levels were significantly increased in HCHWA-D in comparison to the controls, in both frontal and occipital lobes. TGFß-induced pro-fibrotic target genes were also upregulated. We further assessed pathway activation by detecting phospho-SMAD2/3 (pSMAD2/3), a direct TGFß down-stream signaling mediator, using immunohistochemistry. We found abnormal pSMAD2/3 granular deposits specifically on HCHWA-D angiopathic frontal and occipital vessels. We graded pSMAD2/3 accumulation in angiopathic vessels and found a positive correlation with the CAA load independent of the brain area. We also observed pSMAD2/3 granules in a halo surrounding occipital vessels, which was specific for HCHWA-D. The result of this study indicates an upregulation of TGFß1 in HCHWA-D, as was found previously in AD with CAA pathology. We discuss the possible origins and implications of the TGFß pathway deregulation in the microvasculature in HCHWA-D. These findings identify the TGFß pathway as a potential biomarker of disease progression and a possible target of therapeutic intervention in HCHWA-D.


Cerebral Amyloid Angiopathy, Familial/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Aged , Aged, 80 and over , Cerebral Amyloid Angiopathy, Familial/pathology , Female , Frontal Lobe/blood supply , Frontal Lobe/metabolism , Frontal Lobe/pathology , Humans , Male , Middle Aged , Occipital Lobe/blood supply , Occipital Lobe/metabolism , Occipital Lobe/pathology , Phosphorylation , Signal Transduction , Up-Regulation
8.
Nat Commun ; 8(1): 2081, 2017 12 12.
Article En | MEDLINE | ID: mdl-29234026

Protein misfolding and aggregation is increasingly being recognized as a cause of disease. In Alzheimer's disease the amyloid-ß peptide (Aß) misfolds into neurotoxic oligomers and assembles into amyloid fibrils. The Bri2 protein associated with Familial British and Danish dementias contains a BRICHOS domain, which reduces Aß fibrillization as well as neurotoxicity in vitro and in a Drosophila model, but also rescues proteins from irreversible non-fibrillar aggregation. How these different activities are mediated is not known. Here we show that Bri2 BRICHOS monomers potently prevent neuronal network toxicity of Aß, while dimers strongly suppress Aß fibril formation. The dimers assemble into high-molecular-weight oligomers with an apparent two-fold symmetry, which are efficient inhibitors of non-fibrillar protein aggregation. These results indicate that Bri2 BRICHOS affects qualitatively different aspects of protein misfolding and toxicity via different quaternary structures, suggesting a means to generate molecular chaperone diversity.


Amyloid beta-Peptides/metabolism , Cataract/pathology , Cerebellar Ataxia/pathology , Cerebral Amyloid Angiopathy, Familial/pathology , Deafness/pathology , Dementia/pathology , Membrane Glycoproteins/metabolism , Protein Aggregation, Pathological/pathology , Adaptor Proteins, Signal Transducing , Amyloid/metabolism , Amyloid Neuropathies, Familial , Circular Dichroism , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/ultrastructure , Microscopy, Electron, Transmission , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Molecular Chaperones/ultrastructure , Protein Binding , Protein Domains/physiology , Protein Folding , Protein Multimerization/physiology , Recombinant Proteins
9.
J Neurol Neurosurg Psychiatry ; 88(11): 982-994, 2017 11.
Article En | MEDLINE | ID: mdl-28844070

Cerebral amyloid angiopathy (CAA) has never been more relevant. The last 5 years have seen a rapid increase in publications and research in the field, with the development of new biomarkers for the disease, thanks to advances in MRI, amyloid positron emission tomography and cerebrospinal fluid biomarker analysis. The inadvertent development of CAA-like pathology in patients treated with amyloid-beta immunotherapy for Alzheimer's disease has highlighted the importance of establishing how and why CAA develops; without this information, the use of these treatments may be unnecessarily restricted. Our understanding of the clinical and radiological spectrum of CAA has continued to evolve, and there are new insights into the independent impact that CAA has on cognition in the context of ageing and intracerebral haemorrhage, as well as in Alzheimer's and other dementias. While the association between CAA and lobar intracerebral haemorrhage (with its high recurrence risk) is now well recognised, a number of management dilemmas remain, particularly when considering the use of antithrombotics, anticoagulants and statins. The Boston criteria for CAA, in use in one form or another for the last 20 years, are now being reviewed to reflect these new wide-ranging clinical and radiological findings. This review aims to provide a 5-year update on these recent advances, as well as a look towards future directions for CAA research and clinical practice.


Cerebral Amyloid Angiopathy/diagnosis , Aged , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/adverse effects , Amyloid beta-Peptides/therapeutic use , Biomarkers/analysis , Brain/pathology , Cerebral Amyloid Angiopathy/chemically induced , Cerebral Amyloid Angiopathy/pathology , Cerebral Amyloid Angiopathy, Familial/diagnosis , Cerebral Amyloid Angiopathy, Familial/pathology , Humans , Immunotherapy , Neuroimaging
10.
Brain Res ; 1622: 149-62, 2015 Oct 05.
Article En | MEDLINE | ID: mdl-26115583

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.


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
12.
Rev Neurosci ; 25(5): 641-51, 2014.
Article En | MEDLINE | ID: mdl-24870607

Hereditary cerebral hemorrhage with amyloidosis - Dutch type is an autosomal dominant hereditary disease caused by a point mutation in the amyloid precursor protein gene on chromosome 21. The mutation causes an amino acid substitution at codon 693 (E22Q), the 'Dutch mutation'. Amyloid ß, the product after cleavage of the amyloid precursor protein, is secreted into the extracellular space. The Dutch mutation leads to altered amyloid ß cleavage and secretion, enhanced aggregation properties, higher proteolysis resistance, lowered brain efflux transporter affinity, and enhanced cell surfaces binding. All these result in amyloid ß accumulation in cerebral vessel walls, causing cell death and vessel wall integrity loss, making cerebral vessel walls in hereditary cerebral hemorrhage with amyloidosis-Dutch type more prone to rupture and obstruction, leading to hemorrhages and infarcts. Studying the effects of altered amyloid ß metabolism due to mutations like the 'Dutch' provides us with a better understanding of amyloid ß toxicity, also in other amyloid ß diseases like sporadic cerebral amyloid angiopathy and Alzheimer's disease.


Amyloid beta-Peptides/metabolism , Cerebral Amyloid Angiopathy, Familial/metabolism , Plaque, Amyloid/metabolism , Amyloid beta-Peptides/genetics , Cerebral Amyloid Angiopathy, Familial/genetics , Cerebral Amyloid Angiopathy, Familial/pathology , Humans , Mutation , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology , Proteolysis
13.
Exp Gerontol ; 48(2): 109-14, 2013 Feb.
Article En | MEDLINE | ID: mdl-23267843

Accumulation of amyloid-beta (Aß) in brain vessel walls and parenchyma, known as cerebral amyloid angiopathy (CAA) and senile plaques (SPs), respectively, plays a key role in Alzheimer's disease (AD) and hereditary cerebral hemorrhage with amyloidosis of the Dutch type (HCHWA-D) pathogenesis. Although the mechanisms underlying CAA and SP formation remain largely unknown, evidence is mounting that local alterations of the extracellular matrix (ECM) in the brain vessel wall and/or parenchyma play an important role. Lysyl oxidase (LOX, E.C. 1.4.3.13) is an inducible amine oxidase that modulates the ECM by catalyzing the formation of molecular covalent cross-links in ECM proteins. The aim of this study is to investigate the association of LOX with CAA and with classic and diffuse SPs in both AD and HCHWA-D cases. We observed an association of LOX with Aß in CAA and with Aß in both classic and diffuse SPs in AD and HCHWA-D cases. In addition, LOX staining was observed in reactive astrocytes associated with these lesions. We conclude that the ECM modulating enzyme LOX is associated with the Aß-related pathological hallmarks of both AD and HCHWA-D, and that our findings provide additional insights into the mechanisms underlying the formation of these lesions.


Alzheimer Disease/enzymology , Amyloid beta-Peptides/analysis , Astrocytes/enzymology , Cerebral Amyloid Angiopathy, Familial/enzymology , Extracellular Matrix Proteins/analysis , Neocortex/enzymology , Protein-Lysine 6-Oxidase/analysis , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Astrocytes/pathology , Case-Control Studies , Cerebral Amyloid Angiopathy, Familial/pathology , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Male , Middle Aged , Neocortex/pathology
14.
Neurobiol Aging ; 32(10): 1774-83, 2011 Oct.
Article En | MEDLINE | ID: mdl-20015576

By phage display, llama-derived heavy chain antibody fragments were selected from non-immune and immune libraries and tested for their affinity and specificity for beta amyloid by phage-ELISA, immunohistochemistry and surface plasmon resonance. We identified eight distinct heavy chain antibody fragments specific for beta amyloid. While three of them recognized vascular and parenchymal beta amyloid deposits, the remaining five heavy chain antibody fragments recognized vascular beta amyloid specifically, failing to bind to parenchymal beta amyloid. These heavy chain antibody fragments, selected from different libraries, demonstrated differential affinity for different epitopes when used for immunohistochemistry. These observations indicate that the llama heavy chain antibody fragments are the first immunologic probes with the ability to differentiate between parenchymal and vascular beta amyloid aggregates. This indicates that vascular and parenchymal beta amyloid deposits are heterogeneous in epitope presence/availability. The properties of these heavy chain antibody fragments make them potential candidates for use in in vivo differential diagnosis of Alzheimer disease and cerebral amyloid angiopathy. Continued use and characterization of these reagents will be necessary to fully understand the performance of these immunoreagents.


Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Blood Vessels/metabolism , Brain/metabolism , Brain/pathology , Connective Tissue/metabolism , Alzheimer Disease/pathology , Amyloid/immunology , Amyloid beta-Peptides/immunology , Blood Vessels/pathology , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/pathology , Cerebral Amyloid Angiopathy, Familial/metabolism , Cerebral Amyloid Angiopathy, Familial/pathology , Connective Tissue/pathology , Enzyme-Linked Immunosorbent Assay/methods , Epitope Mapping/methods , Epitopes/immunology , Epitopes/metabolism , Humans , Immunoglobulin Variable Region/immunology , Peptide Fragments/chemistry , Peptide Fragments/immunology , Peptide Fragments/metabolism , Peptide Library , Sensitivity and Specificity , Surface Plasmon Resonance/methods
15.
Acta Neuropathol ; 119(2): 189-97, 2010 Feb.
Article En | MEDLINE | ID: mdl-19911184

Stop codon mutations in the gene encoding the prion protein (PRNP) are very rare and have thus far only been described in two patients with prion protein cerebral amyloid angiopathy (PrP-CAA). In this report, we describe the clinical, histopathological and pathological prion protein (PrP(Sc)) characteristics of two Dutch patients carrying novel adjacent stop codon mutations in the C-terminal part of PRNP, resulting in either case in hereditary prion protein amyloidoses, but with strikingly different clinicopathological phenotypes. The patient with the shortest disease duration (27 months) carried a Y226X mutation and showed PrP-CAA without any neurofibrillary lesions, whereas the patient with the longest disease duration (72 months) had a Q227X mutation and showed an unusual Gerstmann-Sträussler-Scheinker disease phenotype with numerous cerebral multicentric amyloid plaques and severe neurofibrillary lesions without PrP-CAA. Western blot analysis in the patient with the Q227X mutation demonstrated the presence of a 7 kDa unglycosylated PrP(Sc) fragment truncated at both the N- and C-terminal ends. Our observations expand the spectrum of clinicopathological phenotypes associated with PRNP mutations and show that a single tyrosine residue difference in the PrP C-terminus may significantly affect the site of amyloid deposition and the overall phenotypic expression of the prion disease. Furthermore, it confirms that the absence of the glycosylphosphatidylinositol anchor in PrP predisposes to amyloid plaque formation.


Brain/pathology , Cerebral Amyloid Angiopathy, Familial/genetics , Gerstmann-Straussler-Scheinker Disease/genetics , PrPSc Proteins/genetics , Prions/genetics , Adult , Base Sequence , Blotting, Western , Cerebral Amyloid Angiopathy, Familial/pathology , Codon, Nonsense , Female , Gerstmann-Straussler-Scheinker Disease/pathology , Humans , Middle Aged , Phenotype , Prion Proteins
16.
FASEB J ; 24(1): 229-41, 2010 Jan.
Article En | MEDLINE | ID: mdl-19770225

Cerebral amyloid angiopathy (CAA) is an age-associated condition and a common finding in Alzheimer's disease in which amyloid-beta (Abeta) vascular deposits are featured in >80% of the cases. Familial Abeta variants bearing substitutions at positions 21-23 are primarily associated with CAA, although they manifest with strikingly different clinical phenotypes: cerebral hemorrhage or dementia. The recently reported Piedmont L34V Abeta mutant, located outside the hot spot 21-23, shows a similar hemorrhagic phenotype, albeit less aggressive than the widely studied Dutch E22Q variant. We monitored the apoptotic events occurring after stimulation of human brain microvascular endothelial and smooth muscle cells with nonfibrillar structures of both variants and wild-type Abeta40. Induction of analogous caspase-mediated mitochondrial pathways was elicited by all peptides, although within different time frames and intensity. Activated pathways were susceptible to pharmacological modulation either through direct inhibition of mitochondrial cytochrome c release or by the action of pan- and pathway-specific caspase inhibitors, giving a clear indication of the independent or synergistic engagement of both extrinsic and intrinsic mechanisms. Structural analyses of the Abeta peptides showed that apoptosis preceded fibril formation, correlating with the presence of oligomers and/or protofibrils. The data support the notion that rare genetic mutations constitute unique paradigms to understand the molecular pathogenesis of CAA.


Amyloid beta-Peptides/genetics , Brain/blood supply , Cerebral Amyloid Angiopathy, Familial/genetics , Cerebral Amyloid Angiopathy, Familial/pathology , Amino Acid Substitution , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Apoptosis , Brain/metabolism , Brain/pathology , Caspases/metabolism , Cell Line , Cerebral Amyloid Angiopathy, Familial/metabolism , Cytochromes c/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Genetic Variation , Humans , Mitochondria/metabolism , Mutation , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
17.
Amyloid ; 15(4): 275-7, 2008 Dec.
Article En | MEDLINE | ID: mdl-19065301

A patient with Down's syndrome (DS) died of cerebral hemorrhage at age 52. At autopsy, a large sub-cortical hematoma was present in the right frontal lobe, and microscopic examination showed numerous senile plaques and neurofibrillary tangles in an extensive area of neocortex and also disclosed heavy involvement of vascular walls by amyloid deposition. These senile plaque and vascular amyloid deposits were specifically stained with an antibody to Abeta. His APOE genotype was epsilon4/epsilon4. This is a rare case of DS with cerebral amyloid angiopathy (CAA)-related cerebral hemorrhage. Genetic factors, such as APOE genotype, conceivably determine the risk of vascular rupture in individuals with CAA, even among patients with DS.


Cerebral Amyloid Angiopathy, Familial/complications , Cerebral Hemorrhage/etiology , Down Syndrome/complications , Amyloid beta-Peptides/genetics , Apolipoproteins E/genetics , Cerebral Amyloid Angiopathy, Familial/genetics , Cerebral Amyloid Angiopathy, Familial/pathology , Cerebral Hemorrhage/pathology , Down Syndrome/genetics , Down Syndrome/pathology , Genotype , Humans , Male , Middle Aged
18.
Am J Pathol ; 171(6): 1989-99, 2007 Dec.
Article En | MEDLINE | ID: mdl-18055545

Inefficient clearance of A beta, caused by impaired blood-brain barrier crossing into the circulation, seems to be a major cause of A beta accumulation in the brain of late-onset Alzheimer's disease patients and hereditary cerebral hemorrhage with amyloidosis Dutch type. We observed association of receptor for advanced glycation end products, CD36, and low-density lipoprotein receptor (LDLR) with cerebral amyloid angiopathy in both Alzheimer's disease and hereditary cerebral hemorrhage with amyloidosis Dutch type brains and increased low-density lipoprotein receptor-related protein-1 (LRP-1) expression by perivascular cells in cerebral amyloid angiopathy. We investigated if these A beta receptors are involved in A beta internalization and in A beta-mediated cell death of human cerebrovascular cells and astrocytes. Expression of both the LRP-1 and LDLR by human brain pericytes and leptomeningeal smooth muscle cells, but not by astrocytes, increased on incubation with A beta. Receptor-associated protein specifically inhibited A beta-mediated up-regulation of LRP-1, but not of LDLR, and receptor-associated protein also decreased A beta internalization and A beta-mediated cell death. We conclude that especially LRP-1 and, to a minor extent, LDLR are involved in A beta internalization by and A beta-mediated cell death of cerebral perivascular cells. Although perivascular cells may adapt their A beta internalization capacity to the levels of A beta present, saturated LRP-1/LDLR-mediated uptake of A beta results in degeneration of perivascular cells.


Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Cerebral Amyloid Angiopathy, Familial/metabolism , Cerebral Amyloid Angiopathy, Familial/pathology , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Aged , Aged, 80 and over , Amyloid beta-Peptides/analysis , Apoptosis , Blood Vessels/metabolism , Blood Vessels/pathology , Blood-Brain Barrier/metabolism , Brain/blood supply , Brain/metabolism , Brain/pathology , CD36 Antigens/metabolism , Female , Humans , Low Density Lipoprotein Receptor-Related Protein-1/analysis , Male , Pericytes/chemistry , Pericytes/metabolism , Pericytes/pathology , Receptors, LDL/analysis , Receptors, LDL/metabolism
19.
J Neurosci Res ; 85(13): 2917-23, 2007 Oct.
Article En | MEDLINE | ID: mdl-17628026

Hereditary cerebral hemorrhage with amyloidosis, Dutch type (HCHWA-D), is an autosomal dominant disorder caused by the Dutch mutation (E693Q) in the beta-amyloid precursor protein. This mutation produces an aberrant amyloid beta (Abeta) species (AbetaE22Q) and causes severe meningocortical vascular Abeta deposition. We analyzed the Abeta composition of the vascular amyloid in the brains of HCHWA-D patients. Immunohistochemistry demonstrated that the vascular amyloid contained both Abeta40 and Abeta42, with a high Abeta40/Abeta42 ratio. In Western blotting of cerebral microvessel fractions isolated from the brains, both wild-type and Dutch-type Abeta40 were observed as major species. Reverse-phase HPLC-mass spectrometric analysis of the fractions revealed both wild-type and Dutch-type Abeta38 as the other main components of the vascular amyloid. Moreover, we detected peaks corresponding to Dutch-type Abeta42 but not to wild-type Abeta42. These results suggest a pathogenic role for the mutant Abeta42 in addition to the mutant Abeta40 in the cerebral amyloid angiopathy of HCHWA-D.


Amyloid beta-Peptides/metabolism , Blood Vessels/metabolism , Cerebellum/pathology , Cerebral Amyloid Angiopathy, Familial/metabolism , Cerebral Hemorrhage/metabolism , Peptide Fragments/metabolism , Adult , Aged , Aged, 80 and over , Amyloid beta-Peptides/genetics , Cerebral Amyloid Angiopathy, Familial/complications , Cerebral Amyloid Angiopathy, Familial/pathology , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/pathology , Chromatography, High Pressure Liquid/methods , Female , Humans , Male , Mass Spectrometry/methods , Middle Aged , Peptide Fragments/genetics
20.
J Neurol Sci ; 257(1-2): 88-96, 2007 Jun 15.
Article En | MEDLINE | ID: mdl-17374542

Two hereditary forms of cerebrovascular amyloidosis, familial British and Danish dementias (FBD and FDD), share striking similarities with Alzheimer's disease (AD) despite structural differences among their amyloid subunits (ABri in FBD, ADan in FDD, and Abeta in AD). Neuropathological lesions in these disorders include neurofibrillary tangles, parenchymal amyloid and pre-amyloid deposits and overwhelming cerebral amyloid angiopathy co-localizing with reactive microglia and multiple amyloid associated proteins including activation products of the complement cascade. Immunohistochemical analysis of FBD and FDD brain lesions unveiled the presence of serum amyloid P-component (SAP) primarily associated with thioflavin positive amyloid deposits in spite of the significant pre-amyloid burden existing in both disorders. Using affinity chromatography and ELISA binding assays we demonstrated specific, calcium-dependent, saturable, high affinity binding interactions between SAP and ABri/ADan peptides, with dissociation constant values in the sub-nanomolar range and within the same order of magnitude as those resulting from the interaction of SAP with Alzheimer's Abeta1-40 and Abeta1-42. The preferential association of SAP with fibrillar amyloid lesions and not with non-fibrillar pre-amyloid deposits is puzzling, suggesting that SAP modulates the assembly and stability of the final fibril rather than participating in the early steps of protein misfolding and oligomerization.


Brain/metabolism , Cerebral Amyloid Angiopathy, Familial/metabolism , Dementia/metabolism , Plaque, Amyloid/metabolism , Serum Amyloid P-Component/metabolism , Adaptor Proteins, Signal Transducing , Aged , Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Benzothiazoles , Biomarkers/metabolism , Brain/pathology , Brain/physiopathology , Cerebral Amyloid Angiopathy, Familial/pathology , Cerebral Amyloid Angiopathy, Familial/physiopathology , Dementia/pathology , Dementia/physiopathology , Denmark , England , Humans , Immunohistochemistry , Membrane Glycoproteins , Membrane Proteins , Neurofibrils/metabolism , Neurofibrils/pathology , Peptide Fragments/metabolism , Plaque, Amyloid/pathology , Protein Subunits/metabolism , Thiazoles/metabolism
...