Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Neurology ; 101(14): 610-620, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37295957

ABSTRACT

The amyloid cascade model of the pathogenesis of Alzheimer disease (AD) is well supported in observational studies. Its therapeutic corollary asserts that removal of amyloid-ß peptide ("amyloid") would provide clinical benefits. After 2 decades of pursuing the strategy of amyloid removal without success, clinical trials of the antiamyloid monoclonal antibody (AAMA) donanemab and a phase 3 clinical trial of lecanemab have reported clinical benefits linked to amyloid removal. Lecanemab (trade name, Leqembi) is the first with published phase 3 trial results. When administered through IV every 2 weeks to patients with elevated brain amyloid and mild cognitive impairment or mild dementia, lecanemab delayed cognitive and functional worsening by approximately 5 months in an 18-month double-blind, placebo-controlled trial. The trial was well conducted, and the results favoring lecanemab were internally consistent. The demonstration that lecanemab treatment delayed clinical progression in persons with mild symptoms due to AD is a major conceptual achievement, but a better appreciation of the magnitude and durability of benefits for individual patients will require extended observations from clinical practice settings. Amyloid-related imaging abnormalities (ARIA) that were largely asymptomatic occurred in approximately 20%, slightly more than half of which were attributable to treatment and the rest to underlying AD-related amyloid angiopathy. Persons who were homozygous for the APOE ε4 allele had greater ARIA risks. Hemorrhagic complications with longer-term lecanemab use need to be better understood. Administration of lecanemab will place unprecedented pressures on dementia care personnel and infrastructure, both of which need to grow exponentially to meet the challenge.


Subject(s)
Alzheimer Disease , Cerebral Amyloid Angiopathy , Humans , Alzheimer Disease/pathology , Cerebral Amyloid Angiopathy/pathology , Amyloid beta-Peptides , Antibodies, Monoclonal/therapeutic use , Patient Care
2.
J Cereb Blood Flow Metab ; 41(5): 1103-1118, 2021 05.
Article in English | MEDLINE | ID: mdl-32791876

ABSTRACT

Diffuse white matter (WM) disease is highly prevalent in elderly with cerebral small vessel disease (cSVD). In humans, cSVD such as cerebral amyloid angiopathy (CAA) often coexists with Alzheimer's disease imposing a significant impediment for characterizing their distinct effects on WM. Here we studied the burden of age-related CAA pathology on WM disease in a novel transgenic rat model of CAA type 1 (rTg-DI). A cohort of rTg-DI and wild-type rats was scanned longitudinally using MRI for characterization of morphometry, cerebral microbleeds (CMB) and WM integrity. In rTg-DI rats, a distinct pattern of WM loss was observed at 9 M and 11 M. MRI also revealed manifestation of small CMB in thalamus at 6 M, which preceded WM loss and progressively enlarged until the moribund disease stage. Histology revealed myelin loss in the corpus callosum and thalamic CMB in all rTg-DI rats, the latter of which manifested in close proximity to occluded and calcified microvessels. The quantitation of CAA load in rTg-DI rats revealed that the most extensive microvascular Aß deposition occurred in the thalamus. For the first time using in vivo MRI, we show that CAA type 1 pathology alone is associated with a distinct pattern of WM loss.


Subject(s)
Brain/blood supply , Cerebral Amyloid Angiopathy/pathology , Cerebral Hemorrhage/pathology , White Matter/pathology , Animals , Brain/diagnostic imaging , Brain/pathology , Calcinosis/complications , Case-Control Studies , Cerebral Amyloid Angiopathy/complications , Cerebral Hemorrhage/diagnostic imaging , Cerebral Small Vessel Diseases/complications , Corpus Callosum/pathology , Diffusion Tensor Imaging/methods , Disease Models, Animal , Female , Global Burden of Disease/statistics & numerical data , Magnetic Resonance Imaging/methods , Male , Microvessels/metabolism , Microvessels/pathology , Rats , Rats, Transgenic , Thalamus/pathology , White Matter/diagnostic imaging
3.
Int J Mol Sci ; 17(1)2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26797603

ABSTRACT

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


Subject(s)
Anticholesteremic Agents/pharmacology , Atorvastatin/pharmacology , Cerebral Amyloid Angiopathy/drug therapy , Cerebral Hemorrhage/drug therapy , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cerebral Amyloid Angiopathy/genetics , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/pathology , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Disease Models, Animal , Gene Expression , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Mice, Transgenic
4.
Stroke ; 46(6): 1633-1640, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25944329

ABSTRACT

BACKGROUND AND PURPOSE: Cerebral amyloid angiopathy (CAA) is a common cause of recurrent intracerebral hemorrhage in the elderly. Previous studies have shown that CAA induces inflammation and expression of matrix metalloproteinase-2 and matrix metalloproteinase-9 (gelatinases) in amyloid-laden vessels. Here, we inhibited both using minocycline in CAA mouse models to determine whether spontaneous intracerebral hemorrhage could be reduced. METHODS: Tg2576 (n=16) and 5xFAD/ApoE4 knockin mice (n=16), aged 17 and 12 months, respectively, were treated with minocycline (50 mg/kg, IP) or saline every other day for 2 months. Brains were extracted and stained with X-34 (to quantify amyloid), Perls' blue (to quantify hemorrhage), and immunostained to examined ß-amyloid peptide load, gliosis (glial fibrillary acidic protein [GFAP], Iba-1), and vascular markers of blood-brain barrier integrity (zonula occludins-1 [ZO-1] and collagen IV). Brain extracts were used to quantify mRNA for a variety of inflammatory genes. RESULTS: Minocycline treatment significantly reduced hemorrhage frequency in the brains of Tg2576 and 5xFAD/ApoE4 mice relative to the saline-treated mice, without affecting CAA load. Gliosis (GFAP and Iba-1 immunostaining), gelatinase activity, and expression of a variety of inflammatory genes (matrix metalloproteinase-9, NOX4, CD45, S-100b, and Iba-1) were also significantly reduced. Higher levels of microvascular tight junction and basal lamina proteins were found in the brains of minocycline-treated Tg2576 mice relative to saline-treated controls. CONCLUSIONS: Minocycline reduced gliosis, inflammatory gene expression, gelatinase activity, and spontaneous hemorrhage in 2 different mouse models of CAA, supporting the importance of matrix metalloproteinase-related and inflammatory pathways in intracerebral hemorrhage pathogenesis. As a Food and Drug Administration-approved drug, minocycline might be considered for clinical trials to test efficacy in preventing CAA-related intracerebral hemorrhage.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cerebral Amyloid Angiopathy/drug therapy , Cerebral Hemorrhage/prevention & control , Minocycline/pharmacology , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cerebral Amyloid Angiopathy/complications , Cerebral Amyloid Angiopathy/genetics , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/pathology , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Leukocyte Common Antigens , Matrix Metalloproteinase 9/biosynthesis , Mice , Mice, Transgenic , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , NADPH Oxidase 4 , NADPH Oxidases/biosynthesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , S100 Calcium Binding Protein beta Subunit/biosynthesis
5.
Aging Cell ; 12(2): 224-36, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23413811

ABSTRACT

Development of cerebral amyloid angiopathy (CAA) and Alzheimer's disease (AD) is associated with failure of elimination of amyloid-ß (Aß) from the brain along perivascular basement membranes that form the pathways for drainage of interstitial fluid and solutes from the brain. In transgenic APP mouse models of AD, the severity of cerebral amyloid angiopathy is greater in the cerebral cortex and hippocampus, intermediate in the thalamus, and least in the striatum. In this study we test the hypothesis that age-related regional variation in (1) vascular basement membranes and (2) perivascular drainage of Aß contribute to the different regional patterns of CAA in the mouse brain. Quantitative electron microscopy of the brains of 2-, 7-, and 23-month-old mice revealed significant age-related thickening of capillary basement membranes in cerebral cortex, hippocampus, and thalamus, but not in the striatum. Results from Western blotting and immunocytochemistry experiments showed a significant reduction in collagen IV in the cortex and hippocampus with age and a reduction in laminin and nidogen 2 in the cortex and striatum. Injection of soluble Aß into the hippocampus or thalamus showed an age-related reduction in perivascular drainage from the hippocampus but not from the thalamus. The results of the study suggest that changes in vascular basement membranes and perivascular drainage with age differ between brain regions, in the mouse, in a manner that may help to explain the differential deposition of Aß in the brain in AD and may facilitate development of improved therapeutic strategies to remove Aß from the brain in AD.


Subject(s)
Aging/pathology , Amyloid beta-Peptides/metabolism , Basement Membrane/pathology , Cerebral Amyloid Angiopathy/pathology , Aging/metabolism , Animals , Basement Membrane/metabolism , Calcium-Binding Proteins , Capillaries/metabolism , Capillaries/pathology , Cell Adhesion Molecules , Cerebral Amyloid Angiopathy/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Collagen Type IV/metabolism , Corpus Striatum/metabolism , Corpus Striatum/pathology , Extracellular Fluid/metabolism , Female , Hippocampus/metabolism , Hippocampus/pathology , Humans , Laminin/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Organ Specificity , Thalamus/metabolism , Thalamus/pathology
6.
Acta Neuropathol ; 118(1): 87-102, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19234858

ABSTRACT

The introduction of immunotherapy and its ultimate success will require re-evaluation of the pathogenesis of Alzheimer's disease particularly with regard to the role of the ageing microvasculature and the effects of APOE genotype. Arteries in the brain have two major functions (a) delivery of blood and (b) elimination of interstitial fluid and solutes, including amyloid-beta (Abeta), along perivascular pathways (lymphatic drainage). Both these functions fail with age and particularly severely in Alzheimer's disease and vascular dementia. Accumulation of Abeta as plaques in brain parenchyma and artery walls as cerebral amyloid angiopathy (CAA) is associated with failure of perivascular elimination of Abeta from the brain in the elderly and in Alzheimer's disease. High levels of soluble Abeta in the brain correlate with cognitive decline in Alzheimer's disease and reflect the failure of perivascular drainage of solutes from the brain and loss of homeostasis of the neuronal environment. Clinically and pathologically, there is a spectrum of disease related to functional failure of the ageing microvasculature with "pure" Alzheimer's disease at one end of the spectrum and vascular dementia at the other end. Changes in the cerebral microvasculature with age have a potential impact on therapy with cholinesterase inhibitors and especially on immunotherapy that removes Abeta from plaques in the brain, but results in an increase in severity of CAA and no clear improvement in cognition. Drainage of Abeta along perivascular pathways in ageing artery walls may need to be improved to maximise the potential for improvement of cognitive function with immunotherapy.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/therapy , Brain/blood supply , Cerebral Amyloid Angiopathy/pathology , Microvessels/pathology , Aging , Alzheimer Disease/complications , Alzheimer Disease/metabolism , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Brain/pathology , Cerebral Amyloid Angiopathy/complications , Cholinesterase Inhibitors/therapeutic use , Dementia, Vascular/pathology , Dementia, Vascular/physiopathology , Humans , Microvessels/physiopathology
7.
J Neuroinflammation ; 4: 22, 2007 Sep 18.
Article in English | MEDLINE | ID: mdl-17877807

ABSTRACT

The deposition of amyloid beta-protein (A beta) in cerebral vasculature, known as cerebral amyloid angiopathy (CAA), is a common pathological feature of Alzheimer's disease and related disorders. In familial forms of CAA single mutations in the A beta peptide have been linked to the increase of vascular A beta deposits accompanied by a strong localized activation of glial cells and elevated expression of neuroinflammatory mediators including complement proteins. We have developed human amyloid-beta precursor protein transgenic mice harboring two CAA A beta mutations (Dutch E693Q and Iowa D694N) that mimic the prevalent cerebral microvascular A beta deposition observed in those patients, and the Swedish mutations (K670N/M671L) to increase A beta production. In these Tg-SwDI mice, we have reported predominant fibrillar A beta along microvessels in the thalamic region and diffuse plaques in cortical region. Concurrently, activated microglia and reactive astrocytes have been detected primarily in association with fibrillar cerebral microvascular A beta in this model. Here we show that three native complement components in classical and alternative complement pathways, C1q, C3, and C4, are elevated in Tg-SwDI mice in regions rich in fibrillar microvascular A beta. Immunohistochemical staining of all three proteins was increased in thalamus, hippocampus, and subiculum, but not frontal cortex. Western blot analysis showed significant increases of all three proteins in the thalamic region (with hippocampus) as well as the cortical region, except C3 that was below detection level in cortex. Also, in the thalamic region (with hippocampus), C1q and C3 mRNAs were significantly up-regulated. These complement proteins appeared to be expressed largely by activated microglial cells associated with the fibrillar microvascular A beta deposits. Our findings demonstrate that Tg-SwDI mice exhibit elevated complement protein expression in response to fibrillar vascular A beta deposition that is observed in patients with familial CAA.


Subject(s)
Amyloid beta-Peptides/genetics , Cerebral Amyloid Angiopathy/metabolism , Complement C1q/metabolism , Complement C3/metabolism , Complement C4/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain/blood supply , Cerebral Amyloid Angiopathy/genetics , Cerebral Amyloid Angiopathy/pathology , Complement C1q/genetics , Complement C3/genetics , Complement C4/genetics , Disease Models, Animal , Hippocampus/blood supply , Hippocampus/metabolism , Hippocampus/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Mutation/genetics , Thalamus/blood supply , Thalamus/metabolism , Thalamus/pathology
8.
Brain ; 129(Pt 11): 2966-76, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16959815

ABSTRACT

Different duplications of the APP locus have been identified in five families with autosomal dominant early onset Alzheimer's disease (ADEOAD) and Abeta-related cerebral amyloid angiopathy (CAA). This study describes the phenotype of this new entity. Clinical, neuropsychological, imagery and neuropathological data were reviewed. The phenotype was not dependent on the size of the duplication and there was no clinical feature of Down's syndrome. Dementia was observed in all cases; intracerebral haemorrhage (ICH) was reported in 6 (26%) and seizures occurred in 12 (57%) of 21 patients. Age of onset of dementia ranged from 42 to 59 years, ICH from 53 to 64 years and age at death from 46 to 75 years. The neuropathological findings in five cases demonstrated Alzheimer's disease and severe CAA lesions that were reminiscent from those reported in brains of Down's syndrome patients. A striking feature consisted in intraneuronal Abetax-40 accumulation located in the granular cell layer of the dentate gyrus and in the pyramidal cell layer of the Ammon's horn.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Cerebral Amyloid Angiopathy/genetics , Gene Duplication , Adult , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Brain/pathology , Cerebral Amyloid Angiopathy/pathology , Cerebral Amyloid Angiopathy/psychology , Cerebral Hemorrhage/etiology , Down Syndrome/genetics , Down Syndrome/pathology , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neuropsychological Tests , Pedigree , Phenotype , Tomography, X-Ray Computed
9.
AJNR Am J Neuroradiol ; 20(4): 637-42, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10319975

ABSTRACT

BACKGROUND AND PURPOSE: Patients with spontaneous intracerebral hemorrhage (ICH) frequently have small areas of signal loss on gradient-echo T2*-weighted MR images, which have been suggested to represent remnants of previous microbleeds. Our aim was to provide histopathologic support for this assumption and to clarify whether the presence and location of microbleeds were associated with microangiopathy. METHODS: We performed MR imaging and correlative histopathologic examination in 11 formalin-fixed brains of patients who had died of an ICH (age range, 45-90 years). RESULTS: Focal areas of signal loss on MR images were noted in seven brains. They were seen in a corticosubcortical location in six brains, in the basal ganglia/thalami in five, and infratentorially in three specimens. Histopathologic examination showed focal hemosiderin deposition in 21 of 34 areas of MR signal loss. No other corresponding abnormalities were found; however, hemosiderin deposits were noted without MR signal changes in two brains. All specimens with MR foci of signal loss showed moderate to severe fibrohyalinosis, and there was additional evidence of amyloid angiopathy in two of those brains. CONCLUSION: Small areas of signal loss on gradient echo T2*-weighted images indicate previous extravasation of blood and are related to bleeding-prone microangiopathy of different origins.


Subject(s)
Brain/blood supply , Cerebral Hemorrhage/pathology , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Aged , Aged, 80 and over , Basal Ganglia/blood supply , Cerebellum/blood supply , Cerebral Amyloid Angiopathy/pathology , Cerebral Cortex/blood supply , Female , Fibrosis , Hemosiderin/analysis , Humans , Hyalin/chemistry , Male , Microcirculation/pathology , Middle Aged , Thalamus/blood supply
10.
Brain Res ; 818(1): 41-50, 1999 Feb 06.
Article in English | MEDLINE | ID: mdl-9914436

ABSTRACT

Morphometry of the cerebellum of 11 subjects who died in the severe, final stage of Alzheimer's disease (AD) and of five age-matched subjects without dementia revealed significant atrophy in the AD group, with a decrease in the volume of the molecular layer by 24% and of the granular layer by 22% in comparison with controls. The 32% decrease in the total number of Purkinje cells that was observed correlates with the atrophy of the molecular layer, whereas the 30% reduction in the total number of granule cells correlates with the atrophy of the molecular and granular layers. A unique pattern of Alzheimer-type pathology was observed in the cerebellum: (1) there were no neurofibrillary changes in the cerebellum of either the control or the AD subjects, (2) there was almost the same extent of leptomeningeal and cortical amyloid angiopathy in the normal aged subjects and in the AD patients, and (3) the presence of plaques was noted in the AD group, but not in the control group. This pattern of pathology suggests that two factors might be considered in the etiopathogenesis of cerebellar atrophy: (1) transneuronal degeneration and neuronal loss resulting from primary pathologic changes in cerebral structures and (2) parenchymal cerebellar ss-amyloidosis. The correlation between the temporal duration of AD and both the decrease of the total number of granule cells (r=0.86, p<0.01) and the volumetric loss of the molecular (r=0.73, p<0.05) and granular (r=0.93, p<0.001) layers of the cerebellar cortex indicates that these cerebellar atrophic changes are likely to be related to the basic pathologic process of AD. Similarly, the correlation between the most complex parameter the atrophy of the cerebellar cortex and the Functional Assessment Staging (FAST) measure of the clinical severity of AD at the time of demise (r=0.63, p<0.05) as well as with the duration of AD (r=0.78, p<0.01) indicates that cerebellar pathology, when viewed holistically, evolves continuously in association with clinical changes throughout the clinically manifest course of AD.


Subject(s)
Alzheimer Disease/pathology , Cerebellum/pathology , Aged , Aged, 80 and over , Atrophy , Case-Control Studies , Cerebral Amyloid Angiopathy/pathology , Cerebral Cortex/pathology , Female , Humans , Male , Meninges/pathology , Neurons/pathology , Purkinje Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL