Abstract
The shared role of amyloid-β (Aβ) deposition in cerebral amyloid angiopathy (CAA) and Alzheimer disease (AD) is arguably the clearest instance of cross-talk between neurodegenerative and cerebrovascular processes. The pathogenic pathways of CAA and AD intersect at the levels of Aβ generation, its circulation within the interstitial fluid and perivascular drainage pathways and its brain clearance, but diverge in their mechanisms of brain injury and disease presentation. Here, we review the evidence for and pathogenic implications of interactions between CAA and AD. Both pathologies seem to be driven by impaired Aβ clearance, creating conditions for a self-reinforcing cycle of increased vascular Aβ, reduced perivascular clearance and further CAA and AD progression. Despite the close relationship between vascular and plaque Aβ deposition, several factors favour one or the other, such as the carboxy-terminal site of the peptide and specific co-deposited proteins. Amyloid-related imaging abnormalities that have been seen in trials of anti-Aβ immunotherapy are another probable intersection between CAA and AD, representing overload of perivascular clearance pathways and the effects of removing Aβ from CAA-positive vessels. The intersections between CAA and AD point to a crucial role for improving vascular function in the treatment of both diseases and indicate the next steps necessary for identifying therapies.
ToC summary
Amyloid-β deposition underlies the pathogenesis of cerebral amyloid angiopathy (CAA) and Alzheimer disease (AD), but the disease pathways differ. Here, Greenberg et al. consider the interactions between CAA and AD, the factors that determine which disease pathway transpires, and the implications for therapeutic development.
Introduction
The contemporary understanding of age-related cognitive impairment centres on the interplay between neurodegenerative disease and cerebrovascular disease. A substantial body of literature suggests that cognitive impairment in the ageing brain is typically driven by overlapping neurodegenerative and cerebrovascular pathologies1. This overlap is demonstrated by findings from the clinical–pathological Religious Orders Study and Memory and Aging Project2, in which Alzheimer disease (AD) pathology was the most prevalent pathology (present in ~65% of brains), but four of the next five most prevalent were vascular pathologies: gross ischaemic infarcts, moderate-to-severe cerebral amyloid angiopathy (CAA), atherosclerosis and arteriolosclerosis. Each of the vascular pathologies was present in >30% of brains. When present, each accounted for a mean of 20–30% of an individual’s age-related cognitive decline. Knowledge that overlap between neurodegenerative and vascular pathologies has a role in cognitive impairment affects prevention strategies. Indeed, over the past 3–4 decades, the age-specific incidence of dementia in developed countries has declined3 in parallel with improvements in blood pressure control and vascular health, suggesting that that these improvements have at least partly reduced the vascular contribution to cognitive impairment and dementia (VCID).
Interactive effects of neurodegenerative and cerebrovascular disease on cognition certainly result from the cumulative brain injuries caused by each process. However, at a more mechanistic level, the two processes might also interact through cross-talk between neurodegenerative pathways and the blood vessels4. The most clear-cut examples of mechanistic interactions are seen in CAA and AD. CAA involves cerebrovascular deposition of amyloid-β (Aβ), which is also the primary constituent of neuritic plaques in AD (Fig. 1). The two conditions often overlap, presumably because of the shared role of Aβ. CAA might also contribute to the pathogenesis of AD by affecting perivascular drainage, a major route of Aβ clearance from the brain5. Given that overlap between sporadic CAA and AD is high6,7, virtually every clinical trial of treatment for sporadic AD or CAA can be considered as a trial for treatment of both, underlining the importance of identifying the interactions between these two processes in disease pathogenesis.
Figure 1 |.
Co-existing amyloid-β in neuritic plaques and vessel walls. Anti-amyloid-β immunostaining (clone 6F/3D, Agilent, 1:200) of a postmortem section of the occipital lobe from a 67-year-old man (left) reveals co-existing neuritic plaques (right, top) and cerebral amyloid angiopathy (CAA; right, bottom) in leptomeningeal and cortical vessels.
In this Review, we summarize the current understanding of the relationship between CAA and AD, how the common factor of Aβ deposition can generate two different disease pathways, how each pathology affects the other, and how the two pathologies together promote cognitive impairment and neurological dysfunction. We focus on the pathological and pathogenic similarities and differences between CAA-related and AD-related brain injury, the central role of perivascular clearance in accumulation of Aβ in the brain, the biochemical and genetic features that favour deposition of Aβ in the vessels or in plaques, and the potential role of CAA in the development of amyloid-related imaging abnormalities (ARIA) that have been identified as major adverse effects of anti-Aβ immunotherapy.
Brain injury in CAA and AD
CAA and AD pathology frequently co-occur in the same brain, presumably because Aβ is pathogenic in both. For example, in an autopsy analysis of 404 brains from individuals in the Religious Orders Study (mean age at death 86.5 ± 7.0 years), the correlation coefficient between CAA severity and AD severity was 0.68 (ref.6). Another autopsy study of 3,976 brains in the US National Alzheimer’s Coordinating Center database (mean age at death 83.2 ± 8.4 years) revealed that the pathological grade of CAA was moderate to severe in 5.3% of brains with no neuritic plaques, 16.1% of brains with mild neuritic plaques, 31.7% of brains with moderate neuritic plaques, and 45.3% of brains with severe neuritic plaques7. Similarly, use of lobar cerebral microbleeds as a neuroimaging marker of CAA8 has shown that the prevalence and incidence of these lesions is increased in symptomatic patients with AD and cognitively normal individuals who are positive for Aβ on PET imaging, suggesting that CAA-related vessel rupture occurs in the presence as well as the absence of AD9. These examples show that, though CAA and AD do not occur in perfect lockstep7, their co-occurrence far exceeds that of other vascular and neurodegenerative pathologies.
The mechanisms by which AD and CAA lead to brain injury largely do not seem to overlap, however. The precise mechanisms of AD-related brain injury remain unclear10, but seem to centre on Aβ-triggered loss of synapses and neurons, normally measured as a loss of cortical tissue, and the development of hyperphosphorylated tau-containing neurofibrillary lesions, which can now be detected with PET11,12,13. Each measure correlates with cognitive performance in Aβ-positive individuals14,15.
By contrast, CAA-related brain injuries seem to arise from blood vessel dysfunction16, either via loss of vessel integrity and haemorrhage or via loss of normal blood supply and ischaemia. CAA-related haemorrhagic lesions include large symptomatic intracerebral haemorrhages, small (mostly asymptomatic) cerebral microbleeds, and bleeding into the cortical sulci (convexity subarachnoid haemorrhage or cortical superficial siderosis). T2*-weighted MRI detects these haemorrhagic markers with high sensitivity, enabling them to form the basis of the Boston Criteria for CAA8. The primary non-haemorrhagic (presumed ischaemic) forms of brain injury in CAA show up as white matter hyperintensities on T2-weighted MRI, structural disconnection measured with diffusion-tensor MRI, and cerebral microinfarcts. These non-haemorrhagic brain injuries, particularly loss of structural connectivity17, seem to correlate most directly with cognitive impairment and slower gait speed in CAA, suggesting that disconnection has a primary role in vascular cognitive impairment and dementia18. A recent investigation of the underlying anatomical basis of reduced structural connectivity in CAA revealed potential contributions from axonal loss, myelin loss and cerebral microinfarcts19; the latter is the most difficult to detect in vivo but is likely to be the most abundant focal CAA-related brain lesion20. Besides overt vascular or perivascular inflammation associated with an autoimmune subtype of CAA21, inflammation does not seem to be a major component in CAA-related microhaemorrhage or microinfarction22.
Despite the broad distinctions between neurodegenerative brain injury in AD and vascular brain injury in CAA, evidence suggests some overlap in pathogenic mechanisms. For example, cerebrovascular dysfunction, which is one of the earliest detectable abnormalities in sporadic and hereditary CAA23–25, has also been identified as an early step in AD pathogenesis26. Conversely, tau deposition is generally not a prominent feature of CAA pathology but has been observed around Aβ-laden vessels in sporadic and hereditary CAA27. In addition, brain atrophy and cortical thinning seem not to be AD-specific neuroimaging markers but also features of cerebrovascular disease, including CAA28. From a clinical standpoint, the presence of advanced CAA in AD is associated with greater cognitive impairment29 and/or faster cognitive decline30, indicating shared contributions to clinical dysfunction.
Pathogenic mechanisms
Aβ clearance pathways
Pathways that are known to be involved in clearing soluble Aβ from the brain include transport across the blood–brain barrier (BBB), phagocytosis, enzymatic degradation and perivascular drainage5. The extent to which each of these pathways contributes to elimination of Aβ from the brain is unclear, but abnormal perivascular drainage of Aβ from interstitial fluid is believed to have a major role in the pathogenesis of CAA and AD.
In studies published in the 1980s and 1990s, injection of tracers into the brains of rats revealed that interstitial fluid drains alongside intracortical arterioles and leptomeningeal arteries and ends up in extracranial lymph nodes31–33. Neuropathological studies of brains from patients with AD have shown that accumulation of Aβ around arteries is fivefold more common than around veins and that Aβ is first deposited at the periphery of arterioles, at the site of putative interstitial fluid drainage pathways34. These observations support the hypothesis that Aβ exits the brain via perivascular pathways and that reduced perivascular clearance (rather than overproduction of Aβ) is a shared pathogenic mechanism in CAA and AD34–36. Further tracer studies of this putative exit route have suggested that fluid gathers around capillaries and leaves the brain along intramural peri-arterial drainage (IPAD) pathways between the smooth muscle cell basement membranes in the tunica media of arterioles and arteries37,38; these pathways match the predominant distribution of Aβ deposition in CAA39.
An alternative proposed route of perivascular drainage of the interstitial fluid is via the glymphatic system40–42. This pathway, which has mainly been described in the context of cerebrospinal fluid (CSF)–interstitial fluid exchange, involves CSF entering the brain alongside arteries, mixing with interstitial fluid and exiting the brain alongside veins. The movement of fluid might depend on the astrocytic water channel aquaporin-4, which serves as the primary channel for convective flow of fluid from the peri-arterial space into the interstitial space and ultimately the peri-venous compartment43–45. Experiments in aquaporin-4 knockout mice and postmortem human brain tissue showed that tracer clearance was reduced in the absence of aquaporin-4 and aquaporin-4 levels were reduced in astrocytic endfeet in the presence of CAA, suggesting that glymphatic clearance can fail in AD and CAA as a result of aquaporin-4 mislocalization and a consequent decrease in Aβ clearance45,46.
Differing findings about the specific patterns of drainage (peri-arterial or peri-venous) in IPAD and the glymphatic system could be attributed to different experimental procedures (reviewed elsewhere47,48, and this remains a topic for ongoing studies38,45,47,49. The two mechanisms could coexist for different types of convective fluid movements. Both models indicate the importance of perivascular drainage pathways in the pathophysiology of CAA and AD, although IPAD better fits the pattern of Aβ deposition observed in vessels in post-mortem human brains.
Despite consensus that fluid drainage from the brain relies on cardiac output37,49, the exact mechanism responsible for perivascular clearance is incompletely understood. Arterial pulsations generated by the heart have been suggested as a potential motive force49–52, but other models have suggested that these small pulsations cannot create the necessary gradient to push fluid towards the pial surface along arteries (or veins)53,54. An alternative is vasomotion generated by larger, spontaneous, low-frequency contractions and dilations of smooth muscle cells55,56, and a role for this motive force is supported by accumulating evidence54,57. Additional studies are needed to unravel the contributions of arterial pulsations and vasomotion58, and advances in contrast-enhanced MRI could improve visualization of clearance pathways in humans in vivo59,60, an important development towards translating experimental observations to the clinical setting.
Interference with perivascular clearance as a result of vascular Aβ deposition in CAA could establish a self-reinforcing cycle of reduced clearance, increased Aβ deposition, and consequent loss of vascular smooth muscle cells (Fig. 2). This self-reinforcing mechanism would exacerbate AD pathology, owing to reduced Aβ clearance, and CAA-related vessel wall breakdown, vascular lesions and tissue injury. In vivo studies have shown that perivascular clearance and evoked vascular reactivity (functional hyperaemia) is reduced in mice with CAA57,61,62. In patients with CAA, reduced evoked vascular reactivity in response to visual stimulation has also been observed, assessed by measurement of the blood-oxygen-level dependent signal in the visual cortex23–25. Further evidence for reduced perivascular clearance in patients with CAA comes from observation of abnormally enlarged perivascular spaces in the subcortical white matter on in vivo MRI63,64, correlating with a high Aβ burden in the overlying cortex65. Furthermore, topographical colocalization of enlarged perivascular spaces and cortical microbleeds has been observed in patients with CAA66.
Figure 2 |.
Impairment of perivascular drainage in cerebral amyloid angiopathy and Alzheimer disease. Healthy perivascular amyloid-β (Aβ) clearance occurs along the walls of arteries and relies on intact vessels and normal vasoactivity (top). Interference of perivascular clearance by cerebral amyloid angiopathy (CAA) could establish a self-reinforcing cycle of Aβ deposition, loss of vascular smooth muscle cells and vasoactivity, and further reduction in clearance (bottom). This self-reinforcing mechanism would exacerbate both Alzheimer disease pathology by reducing Aβ clearance and CAA-related vascular lesions, such as haemorrhages, and tissue injury.
Several vascular pathologies other than CAA also seem to impair perivascular clearance and could be important in the pathophysiology of AD and CAA. For example, impaired clearance was observed in mice after induced transient hypertension49, vessel occlusions or microinfarcts61,67,68, in line with observations that capillary CAA downstream of occluded arterioles was increased in human post-mortem tissue69. Furthermore, the detrimental effects of vascular pathologies on Aβ clearance from the brain are probably exacerbated by ageing, which involves thickening of vessel walls and reductions in vasoactivity68,70,71. Evidence from mouse and human studies suggests that clearance is also affected by disrupted sleep cycles72,73, with sleep deprivation causing increased brain concentrations of Aβ74,75. A proposed explanation for these intriguing findings is that sleep induces increases in interstitial space volume, leading to increased efflux of solutes, including Aβ72. Therefore, prevention of age-related vascular dysfunction and cerebrovascular pathologies at an early age and promotion of healthy sleep habits are plausible approaches to maximizing perivascular clearance and delaying onset of Aβ accumulation in the brain. Similarly, therapeutic targeting of vascular smooth muscle cells is an interesting possibility for promoting healthy perivascular clearance, thereby reducing the risk of AD and cognitive decline with increasing age.
Aβ deposition
Neuropathological evidence shows that Aβ is initially deposited in the neocortex of the brain as CAA and as parenchymal plaques76,77. Parenchymal pathology subsequently expands into the allocortex, thalamus and basal ganglia, whereas vascular Aβ spreads to allocortical areas and the cerebellum76,77. Leptomeningeal and parenchymal blood vessel Aβ deposition usually affects the posterior lobar brain regions (particularly occipital) and rarely affects the deep grey nuclei, white matter and brainstem77. When Aβ deposition occurs in brain capillaries, the condition is classified as CAA type I; CAA without capillary involvement is classified as CAA type II78. CAA type I tends to be widespread, especially in the neocortex and hippocampus77,79. CAA type I has been specifically associated with neuritic plaques and severe AD pathology79,80.
The characteristically patchy and segmental distribution of CAA pathology81 suggests that vascular Aβ preferentially accumulates at sites of initial Aβ deposition, or ‘seeding’. This preferential accumulation has been observed directly in transgenic mice by use of serial in vivo imaging of CAA progression82. More explicit evidence for a prion-like role of Aβ seeds in triggering CAA comes from animal83 and human84,85 studies in which exogenous exposure to Aβ from sources such as pituitary gland extract or a cadaveric graft can trigger early onset CAA. These findings suggest that amyloid seeding and subsequent expansion is a mechanism of initiation and progression of sporadic CAA, though on the basis of anecdotal evidence, exposure to contaminated tissue as a cause of CAA seems to be extremely rare.
Aβ peptide length
Although Aβ is the main component of neuritic plaques and of CAA pathology, the length of Aβ peptides that form the deposits seems to differ between the lesions (Table 1). Peptides that extend to carboxy position 42 (Aβ42) are mainly deposited in neuritic plaques, whereas shorter Aβ40 peptides are the predominant forms deposited in the walls of leptomeningeal and cortical arteries and, occasionally, veins86–88. Vascular deposits also contain Aβ42, but the Aβ40:Aβ42 ratio is higher than that in plaques89. However, the Aβ40:Aβ42 ratio in capillary deposits is lower than in arteries and veins, and is equivalent to that in neuritic plaques89,90.
Table 1 |.
Features associated with vascular or plaque amyloid-β deposition.
Feature | Primarily associated with vascular Aβ deposition | Primarily associated with plaque Aβ deposition | Associated with vascular and plaque Aβ deposition | Refs |
---|---|---|---|---|
Aβ subtype | Aβ40, Aβ36, Aβ37, Aβ38, Aβ39, Aβ41, N3pE-Aβ40 | Aβ42, Aβ43, N3pE-Aβ42(43) | N/A | 86–90 |
88, 97 | ||||
Genetic alterations | APP missense mutations within the Aβ sequence | APP missense mutations that increase Aβ42:Aβ40 ratio | APP missense mutations within the Aβ sequence | 27,100–105 |
(Dutch (Glu693Gln), Italian | (London (Val717Ile) | (Arctic (Glu693Gly), Flemish | 218,219 | |
(Glu693Lys), Iowa | PSEN1 missense mutations | (Ala692Gly)) | 121,123,220, 221 | |
(Asp693Asn), Piedmont | (generally before codon 200) | APP missense mutations that increase Aβ production without increasing Aβ42:Aβ40 ratio | 125–127 | |
(Leu705Val)) | (Swedish (Lys-Met670-671Asn-Leu) | |||
APP duplication or trisomy 21 | ||||
PSEN1 missense mutations | ||||
(generally after codon 200, including Leu282Val, Leu286Pro | ||||
APOE allele | APOE*ε2 (CAA-associated haemorrhagic phenotype) | N/A | APOE*ε4 | 155,156,222,223 |
Co-deposited proteins (selecteda) | Fibrinogen | Heparan sulfate proteoglycans | Apolipoprotein E | 141,150–152 |
Matrix metalloproteainase 9 | Syndecan-1 and syndecan-3 | Apolipoprotein J (clusterin) | 141,150,170, 171,181 | |
Metalloproteinase inhibitor 3) | Heat shock proteins Hsp20 and HspB8 | Apolipoprotein A-I | 140,145 | |
Norrin | α1-antichymotrypsin | Complement proteins C1q, C3c, C4d, C5–9 | 133,141,144 | |
Collagen-α-2(VI) chain | α 2-macroglobulin) | Vitronectin | 150,170 | |
Sushi repeat-containing protein 1 | Intercellular adhesion molecule 1) | Serum amyloid P component | 135,182 | |
Collagenous Alzheimer amyloid plaque component precursor) | Glypican-1 | 134,224,225 | ||
Syndecan-2 | 226–228,136,141,137,141,141,229,142,143,184,185,181,230 | |||
Agrin | 182 | |||
Collagen XVIII | 183 | |||
Heat shock protein HspB2 |
Limited to proteins that co-deposit with Aβ and have been investigated in human brain vascular and parenchymal Aβ deposits. Aβ, amyloid-β; CAA, cerebral amyloid angiopathy; N3pE, pyroglutamate-modified at Glu-3.
The mechanisms that underlie preferential deposition of Aβ40 in vessel walls and Aβ42 in plaques and capillaries have not yet been elucidated. One hypothesis is that vascular Aβ originates from a different source to Aβ in plaques and is generated locally, principally in smooth muscle cells91,92. However, evidence from transgenic mouse models demonstrates that Aβ with a common neuronal origin can cause CAA and parenchymal Aβ plaques93,94, indicating that neuron-derived Aβ can migrate to and accumulate in the vasculature far from its production site. Given that Aβ42 is less soluble and forms fibrils faster than shorter Aβ peptides95, it is more likely to be retained in the parenchyma and initiate insoluble plaque nucleation. By contrast, the more soluble Aβ40 can diffuse along perivascular drainage pathways to accumulate in the walls of vessels. Nevertheless, evidence suggests that Aβ42 is still the first species to be deposited in the vessel wall96. Other Aβ variants, including N-terminal and C-terminal truncated Aβ peptides and species that contain post-translational modifications (Table 1), have also been identified in human brain tissue, though no specific pathogenic role in CAA has yet been identified for any of these variants. Various analytical methods suggest that truncated Aβ species and Aβ that is pyroglutamate-modified at Glu-3 segregate similarly to full-length, unmodified peptides, whereas species that end at position 41 or before are preferentially deposited in vessels88,97.
Genetic influences
Mutations in the APP gene that encodes amyloid precursor protein can also determine the conformation and deposition sites of Aβ (Table 1). Mutations in APP that flank the Aβ coding region generally increase relative or total levels of Aβ42 and therefore predispose to formation of neuritic plaques and phenotypes associated with early onset AD98. By contrast, APP mutations located within or just outside the Aβ coding region induce a clinicopathological phenotype of prominent CAA. Such a mutation causes the autosomal dominant disorder of Dutch-type hereditary CAA (D-CAA, also known as hereditary cerebral haemorrhage with amyloidosis — Dutch type), which is clinically characterized by early-onset recurrent haemorrhagic strokes and dementia99. The cause is a nucleotide change at APP codon 693 that results in a single amino acid substitution (Glu693Gln) at Aβ position 22 (ref.100). In patients with D-CAA, leptomeningeal and cortical blood vessels are affected by severe CAA with diffuse parenchymal Aβ deposits but few or no dense-core plaques101.
Other mutations have also been identified at Aβ position 22, including the Artic mutation (Glu693Gly)102, the Italian (Glu693Lys)103 mutation, and mutations have also been identified at Aβ positions 21, 23 and 34 within the Aβ sequence, — the Flemish (Ala692Gly)104, Iowa (Asp693Asn)27 and Piedmont (Leu705Val)105 mutations, respectively. Each of these variants causes a common neuropathological phenotype characterized by severe CAA, and the Arctic and Flemish mutations also result in parenchymal fibrillar Aβ deposits106,107. Notably, in patients who carry these familial variants, predominant deposition of Aβ40 species is usually seen in cortical and leptomeningeal arteries without an overall increase in Aβ production103,107–109, also the case in sporadic CAA. The clinical spectrum of these familial CAA forms varies, and includes cerebral haemorrhages, ischaemic brain injury and dementia103,105,110,111.
Extensive investigations have been conducted to determine the pathogenic mechanisms that explain the differences in phenotypes induced by the vasculotropic Aβ peptides112–116. This work has shown that the Aβ peptide that contains the Dutch substitution (AβDutch) forms more fibrils in vitro than the wild-type peptide112,117 and assembles on the surface of cerebrovascular smooth muscle cells, where it induces a pathological response, owing the abnormal charge of the peptide113. Results of other studies have shown that AβDutch is more resistant to proteolytic degradation118,119 and less efficiently cleared across the BBB than wild-type Aβ120. Observations from a transgenic mouse model of D-CAA further suggest that the Dutch mutation promotes Aβ deposition in the cerebral vasculature, possibly by increasing the Aβ :Aβ42 ratio94,115 through a currently undefined mechanism.
Increased APP gene dosage also seems to be sufficient to induce CAA; the condition develops with high penetrance and relatively early onset among individuals with APP duplication121,122 or Down syndrome (in which the chromosome that carries AAP is present in three copies)123. In addition, prominent vascular Aβ deposition has been observed in some patients with familial AD who have mutations in the PSEN1 or PSEN2 genes, which encode presenelin-1 and presenelin-2, respectively124–126. Evidence suggests that mutations in PSEN1 at positions beyond codon 200 cause more severe CAA pathology127 and more parieto-occipital white matter hyperintensities on MRI128 than do mutations at positions before codon 200, raising the possibility that particular alterations in presenilin activity favour vascular Aβ pathology.
APOE is the only susceptibility locus for sporadic CAA that has been identified consistently in genetic analyses129,130. In these analyses, CAA has generally been identified according to lobar location of intracerebral haemorrhage rather than by MRI detection of strictly lobar microbleeds, as the requirement for MRI would limit sample size. As a consequence, the results might be affected by diagnostic misclassification. Another approach has been to analyse correlates of CAA pathology in postmortem brains, but two such studies have produced discordant results.
Analysis of 2,807 brains with pathologically graded CAA found an association with APOE but no other loci identified in genome-wide association studies (GWAS) and none of 21 AD-linked loci identified in previous GWAS131. Another candidate-gene analysis of 29 AD risk loci in 256 brains from individuals aged ≥85 years found at least nominal (P <0.05) associations (including results from imputed genotypes) of 20 loci with noncapillary CAA and 15 with capillary CAA; both pathological measures were strongly associated with APOE132. Determining the basis for the differences in these results represents an important question for future large-scale studies.
Co-deposited proteins and apolipoproteins
Aβ that is deposited in neuritic plaques and vessels is accompanied by Aβ-associated proteins, including complement proteins, serum amyloid P component, α1-antichymotrypsin, glycosaminoglycans, matrix metalloproteinase 9 (MMP9) and various apolipoproteins, such as apolipoprotein E, apolipoprotein J (also known as clusterin) and apolipoprotein A-I133–145 (Table 1). When, how and at what concentrations these associated proteins are co-deposited with Aβ remain to be determined, but some of these components can accelerate or inhibit the formation of Aβ fibrils146–150.
Apolipoprotein E is a principal component of both neuritic plaques and CAA151,152 and has been implicated in the pathogenesis of these disorders by evidence that the APOE*ε4 allele is a major risk factor for both AD153,154 and CAA155,156, that APOE*ε4 is associated with total and vascular Aβ levels157 and that APOE* ε2 is a risk factor for haemorrhagic CAA153–155. The APOE*ε2 allele is protective in AD158. The mechanistic basis of these associations might relate to the fact that binding of Aβ to apolipoprotein E modulates clearance of the peptide across the BBB159. Evidence suggests that this process is apolipoprotein E isoform-specific — apolipoprotein E ε4 tends to change the pathway of Aβ clearance from the low-density lipoprotein receptor-related protein 1 (LRP1) pathway to the less efficient very-low-density lipoprotein (VLDL) receptor, whereas apolipoprotein E ε2 and ε3 mediate faster clearance via both LRP1 and VLDL receptor159. Other proposed mechanisms for apolipoprotein E isoform-specific effects on Aβ deposition include direct competition for clearance pathways160, effects on peptide aggregation and fibrillogenesis161, and inhibition of Aβ-induced MMP9 activity162.
The absence of murine apolipoprotein E in a mouse model of cerebral β-amyloidosis delayed parenchymal deposition of fibrillar Aβ and development of CAA163,164, but the expression of human apolipoprotein E in other transgenic mouse lines prevented Aβ accumulation through a mechanism that depended on the apolipoprotein E isoform165,166. Further studies found that expression of mutant amyloid precursor protein and human apolipoprotein E ε4 in mice led to redistribution of Aβ deposition to the cerebral vessels167, and analysis of mouse apolipoprotein E and human apolipoprotein E ε4 in the same transgenic mouse brain showed that mouse apolipoprotein E co-localized with plaques whereas human apolipoprotein E ε4 co-localized with and promoted cortical (though not leptomeningeal) CAA168. In addition to confirming that mouse and human apolipoprotein E differ, the latter study suggests that apolipoprotein E is differentially involved in the formation of plaques or CAA in blood vessels at different locations. Interestingly, APOE genotype seems to influence Aβ deposition in different blood vessel types in the human brain—APOE*ε4 expression is associated with capillary CAA type I, whereas APOE*ε2 expression is associated with CAA type II78,169.
As is the case for apolipoprotien E, apolipoprotein J is co-deposited with fibrillar Aβ in cerebrovascular and parenchymal lesions141,170,171. Apolipoprotein J interacts with Aβ to prevent Aβ aggregation146,172,173, an effect that reportedly depends on the Aβ:apolipoprotein J ratio174. In a study of the APP–presenilin 1 transgenic mouse model, apolipoprotein J deficiency reduced the number of parenchymal Aβ plaques and increased vascular Aβ deposition175. Apolipoprotein J also regulated Aβ clearance from the brain, as previously proposed176, and its absence could impair Aβ clearance across the BBB, leading to its accumulation in perivascular drainage spaces and more CAA pathology175. In other transgenic AD mouse models, apolipoprotein J deficiency decreased parenchymal fibrillary Aβ, but the shift to cerebrovascular Aβ deposition in the absence of apolipoprotein J was not seen177,178.
Quantitative proteomic analysis of isolated neuritic plaques from patients with AD have verified the involvement of proteins previously associated with Aβ and also identified novel components179,180, most of which have not yet been investigated in CAA. Proteomic analysis of vascular amyloid has confirmed the presence and abundance of apolipoprotein E or apolipoprotein J in leptomeningeal and cortical vessels from the brains of patients with CAA150,181,182. Other potentially important proteins that were identified in these studies as being associated with vascular amyloid but not with neuritic plaques include metalloproteinase inhibitor 3181, norrin, collagen α-2 (VI) chain182 and sushi repeat-containing protein 1 (SRPX1)183. Some of these co-localized proteins suggest mechanisms that contribute to vessel injury, such as the ability of SRPX1 to increase Aβ40-induced caspase activity183, but the pathogenic roles of these proteins, if they have any, are unknown. Deposition of fibrinogen — a precursor of fibrin and a principal contributor to haemostasis — has also been detected in CAA-positive vessels in AD184, and the extent of deposition was greatest in people who were homozygotic for APOE*ε4 (ref.185). In transgenic mouse models, depletion of fibrinogen decreased CAA pathology and cognitive decline184, further suggesting a contribution to vascular amyloid deposition.
CAA and ARIA
ARIA, which can be detected with MRI, are vascular abnormalities that have been seen as a consequence of Aβ-targeted immunotherapy in trials in AD186–187. ARIA are classified into two subtypes186: ARIA-E is characterized by fluid attenuation inversion recovery (FLAIR) MRI hyperintensities that are consistent with vasogenic oedema and sulcal effusions with occasional gyral swelling, whereas ARIA-H is characterized by parenchymal microbleeds and superficial haemosiderin deposition in the leptomeninges, seen on T2*-weighted MRI. ARIA-E typically resolves on imaging over the course of weeks, whereas ARIA-H typically remains on subsequent MRIs. The mechanisms that underlies ARIA are not fully understood, but the available evidence suggests that antibody-mediated breakdown of neuritic plaques releases Aβ that is then deposited in vessels, leading to increased CAA, perivascular inflammation, and/or impaired perivascular clearance.
ARIA and amyloid-β clearance
ARIA have now been observed in multiple passive immunotherapy trials, most prominently in trials of antibodies that target N-terminal or conformational forms of Aβ and that have cleared neuritic plaques. In trials of bapineuzumab, gantenerumab and aducanumab, the incidence of ARIA-E was dose-dependent and APOE*ε4-dependent187–188. In the trial of bapineuzumab, the incidence among people without the APOE*ε4 allele reached 14.2% with the highest dose, whereas the incidence among APOE*ε4 carriers was 15.3%, even though these patients received only the lowest dose187. A higher incidence (~40%) of ARIA-E was reported with use of aducanumab and was most common among APOE*ε4 carriers189. Lower rates of ARIA-E have been reported with use of antibodies that target mid-peptide and C-terminal regions of Aβ190–192, possibly because these antibodies tend to bind to monomeric or specific oligomeric Aβ193, thereby mobilizing less Aβ from neuritic plaques than do antibodies against the N-terminus. A low incidence (< 1%) of ARIA-E has been seen in placebo-treated trial participants and in screening populations and observational studies of autosomal dominant and sporadic AD patients; in these contexts, the condition has primarily been observed in APOE*ε4 carriers190,194,195.
Treatment with anti-amyloid immunotherapy is also associated with ARIA-H, though the increased incidence compared with the incidence among people who receive placebo is less pronounced than that for ARIA-E196,188,189. ARIA-H can also occur in the aftermath of ARIA-E196. In the trial of aducanumab, the APOE*ε4 allele was associated with higher rates of co-occurring ARIA-H and ARIA-E (35% at the highest dose)189. A meta-analysis of immunotherapy trials found that treatment was associated with an increased risk of any ARIA and of ARIA-E but not of ARIA-H197. The lack of an association with ARIA-H might reflect the fact that participants with more than four microbleeds are typically excluded from immunotherapy trials and that the rate of spontaneous incident microbleeds in observational studies is substantial198.
Risk factors for ARIA
The risk factor profile for ARIA could provide insight into the underlying pathophysiology. The risk of ARIA-E is dependent on the antibody dose, but whether the risk is driven primarily by the highest antibody concentration reached or the rate of increasing dose is unclear; limited data suggest that dose titration reduces the risk199,200. Multiple studies have demonstrated an association between a higher APOE*ε4 allele dose and a greater risk of ARIA-E and ARIA-H196,201,202. Furthermore, APOE*ε4 carriers are at substantially increased risk of symptomatic ARIA, which can manifest as headache, lethargy, confusion, neuropsychiatric symptoms and, in rare cases, seizures. Symptomatic ARIA occurs in 1–20% of patients with ARIA-E, and this variability depends on the antibody used, the dose and the prevalence of APOE*ε4188,189,201.
Hypertension, hyperlipidaemia and diabetes seem not to be risk factors for ARIA-E203, and no associations have been found between ARIA and baseline biomarkers of brain amyloid on PET, volumetric MRI measures or most CSF biomarkers, although lower baseline CSF levels of Aβ42 in people without APOE*ε4 was associated with a higher risk of ARIA204.
ARIA and CAA
A plausible explanation for ARIA, first proposed on the basis of postmortem studies of people who received the original active immunotherapy AN1792 (refs205), is that solubilization of Aβ overwhelms the capacity for Aβ clearance via the perivascular CSF bulk flow pathways, leading to amyloid deposition in the arterial wall and accelerated development of CAA (Fig. 3). The fact that the number of microbleeds at baseline and the APOE*ε4 allele are risk factors for both ARIA and CAA support this interpretation, as microbleeds suggest the presence of Aβ in vessel walls and the APOE* ε4 allele suggests a high Aβ burden. Autopsy studies of individuals who received immunotherapy have demonstrated that CAA develops to a greater extent in cortical and leptomeningeal vessels (the locations where ARIA occurs) than untreated individuals and revealed treatment-associated concentric vessel wall splitting206, a key element of CAA-associated vasculopathy207. Therapeutic antibodies against Aβ might not only accelerate vascular deposition but also bind to accessible vascular Aβ and consequently further disrupt vessel integrity, contributing to leakage of proteinaceous fluid (ARIA-E) and red blood cells (ARIA-H).
Figure 3 |.
Mechanisms of amyloid-related imaging abnormalities. a | In the normal physiological state, amyloid-β (Aβ) is cleared from the brain in part via perivascular pathways. b | In Alzheimer disease, accumulation of Aβ in brain parenchyma and vessels results in disrupted vascular integrity and impaired clearance. c | Anti-Aβ immunotherapy mobilizes parenchymal Aβ, which moves into already impaired perivascular drainage pathways, and initiates an immune response to vessels. These effects worsen cerebral amyloid angiopathy and render vessels transiently leaky to proteinaceous fluid and blood products, leading to amyloid-related imaging abnormalities (ARIA; ARIA-E and ARIA-H, respectively). d | Limited evidence suggests that with repeated immunization and continued clearance of vascular Aβ, the structural integrity of vessels and the efficiency of perivascular clearance can improve and the risk of ARIA consequently decreases.
Another line of evidence that links ARIA to CAA is the resemblance of ARIA to the syndrome of CAA-related inflammation (CAA-ri). CAA-ri occurs in a subset of patients with CAA, and its clinical presentations, neuroimaging features and association with APOE*ε4 (refs208,209) are similar to those of ARIA-E. CAA-ri involves infiltration of microglia, T cells and Aβ-containing multinucleated giant cells around CAA-positive vessel segments21, suggesting that spontaneous anti-Aβ autoimmune response occurs. In addition, CSF concentrations of autoantibodies against Aβ are increased during the active phase of CAA-ri210, supporting the hypothesis that the process involves an anti-Aβ autoimmune response and that immunotherapy-related ARIA-E is an iatrogenic version of CAA-ri. CAA-ri is typically treated with immunosuppression, which generally improves clinical symptoms and vasogenic oedema208 and leads to normalization of CSF anti-Aβ antibody concentrations210.
The analogy between ARIA and with CAA-ri suggests that repeated or prolonged immunotherapy would increase the inflammatory response, but ARIA seems to decrease with longer treatment, indicating a more complex relationship. Prolonged immunotherapy treatment might lead to a progressively lower plaque burden, which would reduce amyloid mobilization and subsequent deposition in vessel walls, potentially reducing vascular inflammation and risk of ARIA.
Capillary amyloid deposition and BBB alterations are also likely to be involved in ARIA. Ultrastructural analysis of mice treated with anti-Aβ immunotherapy has revealed disruption of the vascular units that comprise the BBB; for example, the normally tight configuration of astrocyte endfeet that envelop endothelial cells is altered as a result of capillary Aβ211. The same study showed that capillary Aβ was associated with local loss of aquaporin 4 channels that affect extracellular–intracellular fluid flux, which could also contribute to ARIA-E. Loss of apolipoprotein J might also disrupt perivascular drainage pathways, which could, as noted above, shift amyloid deposition to the cerebrovasculature175. In an MRI study of APP-overexpressing mice that had been treated with a mouse analogue of bapineuzumab, transient BBB leakage was associated with microbleeds during early treatment, recapitulating some features of ARIA such as the frequent co-occurrence of ARIA-E and ARIA-H; this effect was not seen in saline-treated transgenic mice or immunotherapy-treated wild-type mice212. These results are consistent with a complex model in which ARIA results from mobilization of aggregated Aβ from the neuritic plaques of AD, which leads to vascular and perivascular Aβ deposition (that is, increased CAA), but also concurrent Aβ mobilization from vessels, which leads to disruption of smooth muscle integrity and the BBB.
Vessel recovery after ARIA
Limited evidence suggests that vessels affected by ARIA are capable of some degree of recovery. Postmortem examination of brains from people who received long-term treatment with AN1792 did not reveal vascular smooth muscle cell loss or vessel wall thinning206. Similarly, in transgenic mice that were exposed to an analogue of bapineuzumab for a prolonged period, variance in vascular smooth muscle cell and collagen IV extracellular matrix thickness returned to wild-type levels when treatment was stopped211. These results raise the (still unproven) possibility that morphology and, consequently, perivascular clearance can recover (Fig. 3). In most individuals who are diagnosed with CAA-ri and are treated with immunosuppressive drugs, clinical symptoms improve and MRI signs of vasogenic oedema resolve,208, which further supports the possibility of vessel recovery following anti-Aβ immune response.
Conclusions and future directions
The evidence reviewed above demonstrates that complex interactions at multiple levels exist between CAA and AD pathophysiology. Major commonalities between the conditions are the shared roles of Aβ production, metabolism and convective clearance from the interstitial fluid via the perivascular and intramural pathways. Within these intersecting pathways, multiple factors favour vascular or parenchymal Aβ deposition; these factors include Aβ40:Aβ42 ratio, Aβ mutations, and the presence and composition of apolipoprotein E and apolipoprotein J. The AD and CAA pathways seem to diverge with respect to how they cause tissue injury: AD pathology promotes neuronal and synaptic loss via undefined mechanisms, whereas CAA generates focal tissue lesions via haemorrhagic and ischaemic vascular brain injury.
The interactions between CAA and AD pathophysiology have several implications for treatment. The apparently central role of perivascular drainage suggests that preserving vascular structure and function would improve amyloid clearance and reduce deposition. Candidate approaches to maintaining vascular health and perivascular clearance pathways include reducing mid-life vascular risk factors213 and improving sleep structure73. Effective treatment of CAA might also have the dual benefit of preventing direct CAA-related brain injury and the self-reinforcing cycle of vascular Aβ deposition that worsens Aβ clearance (Fig. 2). Interventions that worsen CAA, however, such as anti-Aβ immunotherapies that mobilize Aβ from neuritic plaques214, might worsen vascular physiology and perivascular clearance, feeding into this self-reinforcing cycle. Clearance of Aβ from plaques into vessels is a possible mechanism for ARIA and for the observation that cerebrovascular reactivity to visual stimulation (without extensive ARIA) worsened in patients with probable CAA who were treated with the anti-Aβ antibody ponezumab192.
These considerations indicate key steps that would substantially accelerate our understanding of the inter-relationship between CAA and AD and the role of CAA–AD crosstalk in human disease. Development of imaging methods that distinguish between plaque and vascular Aβ in living patients would enable identification of the genetic, environmental and treatment factors that favour Aβ deposition in one compartment versus the other. This goal is clearly challenging, given the close structural relationship between the two types of Aβ deposits. In experiments in an APP transgenic mouse model, the phenoxazine derivative resorufin preferentially bound to vascular Aβ215, but this finding has been taken no further in mouse or human studies.
A second key goal for translational research is development of a non-invasive method for reliably measuring clearance of brain interstitial fluid. Indirect approaches, such as the somewhat invasive method of isotope labelling of Aβ216 and PET imaging of ventricular tau217 have suggested that clearance of interstitial fluid is reduced in patients with AD. However, these approaches are limited by the binding of isotope labeled-Aβ and the tau tracer THK5117 within the brain — an ideal ligand would exit the interstitial fluid via perivascular drainage pathways without binding or uptake of any sort. If these challenges can be overcome, measurement of interstitial fluid clearance would enable testing of candidate treatments for slowing CAA and AD progression and possibly that of other brain diseases associated with impaired solute clearance.
Key Points.
Amyloid-β (Aβ) in the brain interstitial fluid can be cleared via perivascular drainage pathways or deposited as neuritic plaques in the brain parenchyma or as cerebral amyloid angiopathy (CAA) along vessel walls.
Vascular dysfunction caused by CAA reduces perivascular Aβ clearance in animal models, creating a vicious cycle of vascular and parenchymal Aβ accumulation.
Factors that favour vascular Aβ deposition over parenchymal deposition include termination of Aβ at or before position 41, missense mutations within the Aβ coding region, and some co-deposited proteins, such as fibrinogen.
Amyloid-related imaging abnormalities observed in trials of anti-Aβ immunotherapy, might result from mobilization of plaque Aβ into the perivascular drainage system or from antibody targeting of vascular Aβ deposits.
Development of methods for imaging perivascular drainage in humans would be a key step towards identifying treatments for enhancing Aβ clearance and reducing vascular and parenchymal deposition.
Acknowledgments
S.M.G. acknowledges research funding from the National Institutes of Health (R01 NS096730, R01 AG26484, U24 NS100591)
Footnotes
Competing interests
S.M.G. has served as a consultant or safety monitor for Alzheimer immunotherapy trials for Biogen, DIAN-TU and Roche. R.S. has served as a consultant or received clinical research funding from Biogen, Eisai, Eli Lilly, Janssen, Roche and Takeda.
Publisher's note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Peer review information
Nature Reviews Neurology thanks J. Nicoll, M. Verbeek and M. Yamada for their contribution to the peer review of this work.
References
- 1.Corriveau RA et al. The Science of Vascular Contributions to Cognitive Impairment and Dementia (VCID): A Framework for Advancing Research Priorities in the Cerebrovascular Biology of Cognitive Decline. Cell Mol Neurobiol 36, 281–288 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Boyle PA et al. Person-specific contribution of neuropathologies to cognitive loss in old age. Ann. Neurol 83, 74–83 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Satizabal CL et al. Incidence of Dementia over Three Decades in th Framingham Heart Study. N. Engl. J. Med 374, 523–532 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Iadecola C & Gottesman RF Cerebrovascular Alterations in Alzheimer Disease. Circ Res 123, 406–408 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Tarasoff-Conway JM et al. Clearance systems in the brain-implications for Alzheimer disease. Nat Rev Neurol 11, 457–470 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Arvanitakis Z et al. Cerebral amyloid angiopathy pathology and cognitive domains in older persons. Ann. Neurol 69, 320–327 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Brenowitz WD, Nelson PT, Besser LM, Heller KB & Kukull WA Cerebral amyloid angiopathy and its co-occurrence with Alzheimer's disease and other cerebrovascular neuropathologic changes. Neurobiol. Aging 36, 2702–2708 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Greenberg SM & Charidimou A Diagnosis of Cerebral Amyloid Angiopathy: Evolution of the Boston Criteria. Stroke 49, 491–497 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Yates PA et al. Incidence of cerebral microbleeds in preclinical Alzheimer disease. Neurology 82, 1266–1273 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.De Strooper B & Karran E The Cellular Phase of Alzheimer's Disease. Cell 164, 603–615 (2016). [DOI] [PubMed] [Google Scholar]
- 11.Guillozet AL, Weintraub S, Mash DC & Mesulam MM Neurofibrillary tangles, amyloid, and memory in aging and mild cognitive impairment. Arch. Neurol 60, 729–736 (2003). [DOI] [PubMed] [Google Scholar]
- 12.Andrade-Moraes CH et al. Cell number changes in Alzheimer's disease relate to dementia, not to plaques and tangles. Brain 136, 3738–3752 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Villemagne VL & Okamura N Tau imaging in the study of ageing, Alzheimer's disease, and other neurodegenerative conditions. Curr Opin Neurobiol 36, 43–51 (2016). [DOI] [PubMed] [Google Scholar]
- 14.Villeneuve S et al. Cortical thickness mediates the effect of beta-amyloid on episodic memory. Neurology 82, 761–767 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Sperling RA et al. The impact of amyloid-beta and tau on prospective cognitive decline in older individuals. Ann. Neurol 85, 181–193 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Greenberg SM et al. Outcome markers for clinical trials in cerebral amyloid angiopathy. Lancet Neurol 13, 419–428 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Reijmer YD et al. Structural network alterations and neurological dysfunction in cerebral amyloid angiopathy. Brain 138, 179–188 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Dichgans M & Leys D Vascular Cognitive Impairment. Circ Res 120, 573–591 (2017). [DOI] [PubMed] [Google Scholar]
- 19.van Veluw SJ et al. Histopathology of diffusion imaging abnormalities in cerebral amyloid angiopathy. Neurology 92, e933–e943 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.van Veluw SJ et al. Detection, risk factors, and functional consequences of cerebral microinfarcts. Lancet Neurol 16, 730–740 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Eng JA, Frosch MP, Choi K, Rebeck GW & Greenberg SM Clinical manifestations of cerebral amyloid angiopathy-related inflammation. Ann. Neurol 55, 250–256 (2004). [DOI] [PubMed] [Google Scholar]
- 22.van Veluw SJ et al. Different microvascular alterations underlie microbleeds and microinfarcts. Ann. Neurol 86, 279–292 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Dumas A et al. Functional magnetic resonance imaging detection of vascular reactivity in cerebral amyloid angiopathy. Ann. Neurol 72, 76–81 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study and references 24 and 25 demonstrate impairment of vascular reactivity to physiological stimulation in CAA, a mechanism that may contribute to tissue injury and delayed perivascular clearance.
- 24.Peca S et al. Neurovascular decoupling is associated with severity of cerebral amyloid angiopathy. Neurology 81, 1659–1665 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.van Opstal AM et al. Cerebrovascular function in presymptomatic and symptomatic individuals with hereditary cerebral amyloid angiopathy: a case-control study. Lancet Neurol 16, 115–122 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Iturria-Medina Y et al. Early role of vascular dysregulation on late-onset Alzheimer's disease based on multifactorial data-driven analysis. Nat Commun 7, 11934 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Grabowski TJ, Cho HS, Vonsattel JP, Rebeck GW & Greenberg SM Novel amyloid precursor protein mutation in an Iowa family with dementia and severe cerebral amyloid angiopathy. Ann. Neurol 49, 697–705 (2001). [DOI] [PubMed] [Google Scholar]
- 28.Fotiadis P et al. Cortical atrophy in patients with cerebral amyloid angiopathy: a case-control study. Lancet Neurol 15, 811–819 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Goos JD et al. Patients with Alzheimer disease with multiple microbleeds: relation with cerebrospinal fluid biomarkers and cognition. Stroke 40, 3455–3460 (2009). [DOI] [PubMed] [Google Scholar]
- 30.Boyle PA et al. Cerebral amyloid angiopathy and cognitive outcomes in community-based older persons. Neurology 85, 1930–1936 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Szentistvanyi I, Patlak CS, Ellis RA & Cserr HF Drainage of interstitial fluid from different regions of rat brain. Am. J. Physiol 246, F835–844 (1984). [DOI] [PubMed] [Google Scholar]
- 32.Zhang ET, Richards HK, Kida S & Weller RO Directional and compartmentalised drainage of interstitial fluid and cerebrospinal fluid from the rat brain. Acta Neuropathol. 83, 233–239 (1992). [DOI] [PubMed] [Google Scholar]
- 33.Kida S, Pantazis A & Weller RO CSF drains directly from the subarachnoid space into nasal lymphatics in the rat. Anatomy, histology and immunological significance. Neuropathol Appl Neurobiol 19, 480–488 (1993). [DOI] [PubMed] [Google Scholar]
- 34.Weller RO et al. Cerebral amyloid angiopathy: amyloid beta accumulates in putative interstitial fluid drainage pathways in Alzheimer's disease. The American journal of pathology 153, 725–733 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study was an early statement of the experimental support for a link between cCAA and the perivascular fluid drainage process.
- 35.Weller RO, Subash M, Preston SD, Mazanti I & Carare RO Perivascular drainage of amyloid-beta peptides from the brain and its failure in cerebral amyloid angiopathy and Alzheimer's disease. Brain Pathol 18, 253–266 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Carare RO, Hawkes CA, Jeffrey M, Kalaria RN & Weller RO Review: cerebral amyloid angiopathy, prion angiopathy, CADASIL and the spectrum of protein elimination failure angiopathies (PEFA) in neurodegenerative disease with a focus on therapy. Neuropathol Appl Neurobiol 39, 593–611 (2013). [DOI] [PubMed] [Google Scholar]
- 37.Carare RO et al. Solutes, but not cells, drain from the brain parenchyma along basement membranes of capillaries and arteries: significance for cerebral amyloid angiopathy and neuroimmunology. Neuropathol Appl Neurobiol 34, 131–144 (2008). [DOI] [PubMed] [Google Scholar]
- 38.Albargothy NJ et al. Convective influx/glymphatic system: tracers injected into the CSF enter and leave the brain along separate periarterial basement membrane pathways. Acta Neuropathol. 136, 139–152 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Keable A et al. Deposition of amyloid beta in the walls of human leptomeningeal arteries in relation to perivascular drainage pathways in cerebral amyloid angiopathy. Biochim Biophys Acta 1862, 1037–1046 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Iliff JJ et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med 4, 147ra111 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Plog BA & Nedergaard M The Glymphatic System in Central Nervous System Health and Disease: Past, Present, and Future. Annu Rev Pathol 13, 379–394 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Benveniste H et al. The Glymphatic System and Waste Clearance with Brain Aging: A Review. Gerontology 65, 106–119 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Nedergaard M Neuroscience. Garbage truck of the brain. Science 340, 1529–1530 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Smith AJ, Yao X, Dix JA, Jin BJ & Verkman AS Test of the 'glymphatic' hypothesis demonstrates diffusive and aquaporin-4-independent solute transport in rodent brain parenchyma. Elife 6, e27679 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Mestre H et al. Aquaporin-4-dependent glymphatic solute transport in the rodent brain. Elife 7, e40070 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Wilcock DM, Vitek MP & Colton CA Vascular amyloid alters astrocytic water and potassium channels in mouse models and humans with Alzheimer's disease. Neuroscience 159, 1055–1069 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Bakker EN et al. Lymphatic Clearance of the Brain: Perivascular, Paravascular and Significance for Neurodegenerative Diseases. Cell Mol Neurobiol 36, 181–194 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Smith AJ & Verkman AS The “glymphatic” mechanism for solute clearance in Alzheimer's disease: game changer or unproven speculation? FASEB J. 32, 543–551 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Mestre H et al. Flow of cerebrospinal fluid is driven by arterial pulsations and is reduced in hypertension. Nat Commun 9, 4878 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Schley D, Carare-Nnadi R, Please CP, Perry VH & Weller RO Mechanisms to explain the reverse perivascular transport of solutes out of the brain. J Theor Biol 238, 962–974 (2006). [DOI] [PubMed] [Google Scholar]
- 51.Weller RO, Djuanda E, Yow HY & Carare RO Lymphatic drainage of the brain and the pathophysiology of neurological disease. Acta Neuropathol. 117, 1–14 (2009). [DOI] [PubMed] [Google Scholar]
- 52.Bedussi B, Almasian M, de Vos J, VanBavel E & Bakker EN Paravascular spaces at the brain surface: Low resistance pathways for cerebrospinal fluid flow. J Cereb Blood Flow Metab 38, 719–726 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Diem AK et al. Arterial Pulsations cannot Drive Intramural Periarterial Drainage: Significance for Abeta Drainage. Front Neurosci 11, 475 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Aldea R, Weller RO, Wilcock DM, Carare RO & Richardson G Cerebrovascular Smooth Muscle Cells as the Drivers of Intramural Periarterial Drainage of the Brain. Front Aging Neurosci 11, 1 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Mateo C, Knutsen PM, Tsai PS, Shih AY & Kleinfeld D Entrainment of Arteriole Vasomotor Fluctuations by Neural Activity Is a Basis of Blood-Oxygenation-Level-Dependent “Resting-State” Connectivity. Neuron 96, 936–948 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study identified a link between resting neuronal activity and vasomotion, the proposed motor force for perivascular clearance.
- 56.He Y et al. Ultra-Slow Single-Vessel BOLD and CBV-Based fMRI Spatiotemporal Dynamics and Their Correlation with Neuronal Intracellular Calcium Signals. Neuron 97, 925–939 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.van Veluw SJ et al. Vasoactivity as the driving force for paravascular clearance in the awake Alzheimer model mouse brain. 2018 Neuroscience Meeting Planner Program No. 627.03, Online (2018). [Google Scholar]
- 58.Geurts LJ, Zwanenburg JJM, Klijn CJM, Luijten PR & Biessels GJ Higher Pulsatility in Cerebral Perforating Arteries in Patients With Small Vessel Disease Related Stroke, a 7T MRI Study. Stroke 10.1161/STROKEAHA.118.022516 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Eide PK & Ringstad G MRI with intrathecal MRI gadolinium contrast medium administration: a possible method to assess glymphatic function in human brain. Acta Radiol Open 4, 2058460115609635 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Ringstad G, Vatnehol SAS & Eide PK Glymphatic MRI in idiopathic normal pressure hydrocephalus. Brain 140, 2691–2705 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Arbel-Ornath M et al. Interstitial fluid drainage is impaired in ischemic stroke and Alzheimer's disease mouse models. Acta Neuropathol. 126, 353–364 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Xu Z et al. Deletion of aquaporin-4 in APP/PS1 mice exacerbates brain Abeta accumulation and memory deficits. Mol Neurodegener 10, 58 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wardlaw JM, Smith C & Dichgans M Mechanisms of sporadic cerebral small vessel disease: insights from neuroimaging. Lancet Neurol 12, 483–497 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Charidimou A et al. MRI-visible perivascular spaces in cerebral amyloid angiopathy and hypertensive arteriopathy. Neurology 88, 1157–1164 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.van Veluw SJ et al. Cerebral amyloid angiopathy severity is linked to dilation of juxtacortical perivascular spaces. J Cereb Blood Flow Metab 36, 576–580 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Bouvy WH et al. Microbleeds colocalize with enlarged juxtacortical perivascular spaces in amnestic mild cognitive impairment and early Alzheimer’s disease: A 7 Tesla MRI study. J Cerebr Blood F Met 10.1177/0271678X19838087 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Garcia-Alloza M et al. Cerebrovascular lesions induce transient beta-amyloid deposition. Brain 134, 3697–3707 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wang M et al. Focal Solute Trapping and Global Glymphatic Pathway Impairment in a Murine Model of Multiple Microinfarcts. J. Neurosci 37, 2870–2877 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Weller RO, Yow HY, Preston SD, Mazanti I & Nicoll JA Cerebrovascular disease is a major factor in the failure of elimination of Abeta from the aging human brain: implications for therapy of Alzheimer's disease. Ann. N. Y. Acad. Sci 977, 162–168 (2002). [DOI] [PubMed] [Google Scholar]
- 70.Hawkes CA et al. Regional differences in the morphological and functional effects of aging on cerebral basement membranes and perivascular drainage of amyloid-beta from the mouse brain. Aging Cell 12, 224–236 (2013). [DOI] [PubMed] [Google Scholar]
- 71.Kress BT et al. Impairment of paravascular clearance pathways in the aging brain. Ann. Neurol 76, 845–861 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Xie L et al. Sleep drives metabolite clearance from the adult brain. Science 342, 373–377 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Holth JK et al. The sleep-wake cycle regulates brain interstitial fluid tau in mice and CSF tau in humans. Science 363, 880–884 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Kang JE et al. Amyloid-beta dynamics are regulated by orexin and the sleep-wake cycle. Science 326, 1005–1007 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Shokri-Kojori E et al. beta-Amyloid accumulation in the human brain after one night of sleep deprivation. Proc Natl Acad Sci U S A 115, 4483–4488 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Thal DR, Rub U, Orantes M & Braak H Phases of A beta-deposition in the human brain and its relevance for the development of AD. Neurology 58, 1791–1800 (2002). [DOI] [PubMed] [Google Scholar]
- 77.Thal DR, Ghebremedhin E, Orantes M & Wiestler OD Vascular pathology in Alzheimer disease: correlation of cerebral amyloid angiopathy and arteriosclerosis/lipohyalinosis with cognitive decline. J Neuropathol. Exp. Neurol 62, 1287–1301 (2003). [DOI] [PubMed] [Google Scholar]
- 78.Thal DR et al. Two types of sporadic cerebral amyloid angiopathy. J Neuropathol. Exp. Neurol 61, 282–293 (2002). [DOI] [PubMed] [Google Scholar]
- 79.Makela M, Paetau A, Polvikoski T, Myllykangas L & Tanskanen M Capillary amyloid-beta protein deposition in a population-based study (Vantaa 85+). J Alzheimers Dis 49, 149–157 (2016). [DOI] [PubMed] [Google Scholar]
- 80.Attems J & Jellinger KA Only cerebral capillary amyloid angiopathy correlates with Alzheimer pathology--a pilot study. Acta Neuropathol (Berl) 107, 83–90 (2004). [DOI] [PubMed] [Google Scholar]
- 81.Vonsattel JP et al. Cerebral amyloid angiopathy without and with cerebral hemorrhages: a comparative histological study. Ann. Neurol 30, 637–649 (1991). [DOI] [PubMed] [Google Scholar]
- 82.Robbins EM et al. Kinetics of cerebral amyloid angiopathy progression in a transgenic mouse model of Alzheimer disease. J. Neurosci 26, 365–371 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Meyer-Luehmann M et al. Exogenous induction of cerebral beta-amyloidogenesis is governed by agent and host. Science 313, 1781–1784 (2006). [DOI] [PubMed] [Google Scholar]
- 84.Jaunmuktane Z et al. Evidence for human transmission of amyloid-beta pathology and cerebral amyloid angiopathy. Nature 525, 247–250 (2015). [DOI] [PubMed] [Google Scholar]; This initial report of human tissue transmitting early-onset CAA provides strong evidence for amyloid seeding as a mechanism of disease initiation.
- 85.Banerjee G et al. Early onset cerebral amyloid angiopathy following childhood exposure to cadaveric dura. Ann. Neurol 85, 284–290 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Miller DL et al. Peptide compositions of the cerebrovascular and senile plaque core amyloid deposits of Alzheimer's disease. Archives of Biochemistry & Biophysics 301, 41–52 (1993). [DOI] [PubMed] [Google Scholar]
- 87.Gravina SA et al. Amyloid beta protein (A beta) in Alzheimer's disease brain. Biochemical and immunocytochemical analysis with antibodies specific for forms ending at A beta 40 or A beta 42(43). J. Biol. Chem 270, 7013–7016 (1995). [DOI] [PubMed] [Google Scholar]
- 88.Kakuda N et al. Distinct deposition of amyloid-beta species in brains with Alzheimer's disease pathology visualized with MALDI imaging mass spectrometry. Acta Neuropathol Commun 5, 73 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Roher AE et al. beta-Amyloid-(1–42) is a major component of cerebrovascular amyloid deposits: implications for the pathology of Alzheimer disease. Proc Natl Acad Sci U S A 90, 10836–10840 (1993). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Attems J, Lintner F & Jellinger KA Amyloid beta peptide 1–42 highly correlates with capillary cerebral amyloid angiopathy and Alzheimer disease pathology. Acta Neuropathol (Berl) (2004). [DOI] [PubMed] [Google Scholar]
- 91.Wisniewski HM & Wegiel J Beta-amyloid formation by myocytes of leptomeningeal vessels. Acta Neuropathol. 87, 233–241 (1994). [DOI] [PubMed] [Google Scholar]
- 92.Frackowiak J, Miller DL, Potempska A, Sukontasup T & Mazur-Kolecka B Secretion and accumulation of Abeta by brain vascular smooth muscle cells from AbetaPP-Swedish transgenic mice. J Neuropathol. Exp. Neurol 62, 685–696 (2003). [DOI] [PubMed] [Google Scholar]
- 93.Calhoun ME et al. Neuronal overexpression of mutant amyloid precursor protein results in prominent deposition of cerebrovascular amyloid. Proc Natl Acad Sci U S A 96, 14088–14093 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]; This demonstration of CAA pathology in transgenic mice with neuronal expression of mutant amyloid precursor protein helped to establish the principle that neuronally derived amyloid-β could reach sites of deposition in vessel walls.
- 94.Herzig MC et al. Abeta is targeted to the vasculature in a mouse model of hereditary cerebral hemorrhage with amyloidosis. Nat Neurosci 7, 954–960 (2004). [DOI] [PubMed] [Google Scholar]
- 95.Jarrett JT, Berger EP & Lansbury PT The carboxy terminus of the beta amyloid protein is critical for the seeding of amyloid formation: implications for the pathogenesis of Alzheimer's disease. Biochem. 32, 4693–4697 (1993). [DOI] [PubMed] [Google Scholar]
- 96.McGowan E et al. Abeta42 is essential for parenchymal and vascular amyloid deposition in mice. Neuron 47, 191–199 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Harigaya Y et al. Amyloid beta protein starting pyroglutamate at position 3 is a major component of the amyloid deposits in the Alzheimer's disease brain. Biochem. Biophys. Res. Commun 276, 422–427 (2000). [DOI] [PubMed] [Google Scholar]
- 98.Revesz T et al. Cerebral amyloid angiopathies: a pathologic, biochemical, and genetic view. J Neuropathol. Exp. Neurol 62, 885–898 (2003). [DOI] [PubMed] [Google Scholar]
- 99.Wattendorff AR, Frangione B, Luyendijk W & Bots GT Hereditary cerebral haemorrhage with amyloidosis, Dutch type (HCHWA-D): clinicopathological studies. J. Neurol. Neurosurg. Psychiatry 58, 699–705 (1995). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Levy E et al. Mutation of the Alzheimer's disease amyloid gene in hereditary cerebral hemorrhage, Dutch type. Science 248, 1124–1126 (1990). [DOI] [PubMed] [Google Scholar]
- 101.Maat-Schieman ML, van Duinen SG, Bornebroek M, Haan J & Roos RA Hereditary cerebral hemorrhage with amyloidosis-Dutch type (HCHWA-D): II--A review of histopathological aspects. Brain pathology 6, 115–120 (1996). [DOI] [PubMed] [Google Scholar]
- 102.Kamino K et al. Linkage and mutational analysis of familial Alzheimer disease kindreds for the APP gene region. Am. J. Hum. Genet 51, 998–1014 (1992). [PMC free article] [PubMed] [Google Scholar]
- 103.Bugiani O et al. Hereditary cerebral hemorrhage with amyloidosis associated with the E693K mutation of APP. Arch. Neurol 67, 987–995 (2010). [DOI] [PubMed] [Google Scholar]
- 104.Cras P et al. Presenile Alzheimer dementia characterized by amyloid angiopathy and large amyloid core type senile plaques in the APP 692Ala-->Gly mutation. Acta Neuropathol. 96, 253–260 (1998). [DOI] [PubMed] [Google Scholar]
- 105.Obici L et al. A novel AbetaPP mutation exclusively associated with cerebral amyloid angiopathy. Ann. Neurol 58, 639–644 (2005). [DOI] [PubMed] [Google Scholar]
- 106.Basun H et al. Clinical and neuropathological features of the arctic APP gene mutation causing early-onset Alzheimer disease. Arch. Neurol 65, 499–505 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Kumar-Singh S et al. Dense-core senile plaques in the Flemish variant of Alzheimer's disease are vasocentric. Am. J. Pathol 161, 507–520 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Mann DM et al. Predominant deposition of amyloid-beta 42(43) in plaques in cases of Alzheimer's disease and hereditary cerebral hemorrhage associated with mutations in the amyloid precursor protein gene. The American journal of pathology 148, 1257–1266 (1996). [PMC free article] [PubMed] [Google Scholar]
- 109.Shin Y et al. Abeta species, including IsoAsp23 Abeta, in Iowa-type familial cerebral amyloid angiopathy. Acta Neuropathol. 105, 252–258 (2003). [DOI] [PubMed] [Google Scholar]
- 110.Greenberg SM et al. Hemorrhagic stroke associated with the Iowa amyloid precursor protein mutation. Neurology 60, 1020–1022 (2003). [DOI] [PubMed] [Google Scholar]
- 111.Brooks WS et al. Hemorrhage is uncommon in new Alzheimer family with Flemish amyloid precursor protein mutation. Neurology 63, 1613–1617 (2004). [DOI] [PubMed] [Google Scholar]
- 112.Clements A, Walsh DM, Williams CH & Allsop D Effects of the mutations Glu22 to Gln and Ala21 to Gly on the aggregation of a synthetic fragment of the Alzheimer's amyloid beta/A4 peptide. Neurosci. Lett 161, 17–20 (1993). [DOI] [PubMed] [Google Scholar]
- 113.Melchor JP, McVoy L & Van Nostrand WE Charge alterations of E22 enhance the pathogenic properties of the amyloid beta-protein. J. Neurochem 74, 2209–2212 (2000). [DOI] [PubMed] [Google Scholar]
- 114.Van Nostrand WE, Melchor JP, Romanov G, Zeigler K & Davis J Pathogenic effects of cerebral amyloid angiopathy mutations in the amyloid beta-protein precursor. Ann. N. Y. Acad. Sci 977, 258–265 (2002). [DOI] [PubMed] [Google Scholar]
- 115.Nilsberth C et al. The 'Arctic' APP mutation (E693G) causes Alzheimer's disease by enhanced Abeta protofibril formation. Nat Neurosci 4, 887–893 (2001). [DOI] [PubMed] [Google Scholar]
- 116.Fossati S et al. Differential activation of mitochondrial apoptotic pathways by vasculotropic amyloid-beta variants in cells composing the cerebral vessel walls. FASEB J. 24, 229–241 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Miravalle L et al. Substitutions at codon 22 of Alzheimer's abeta peptide induce diverse conformational changes and apoptotic effects in human cerebral endothelial cells. J. Biol. Chem 275, 27110–27116 (2000). [DOI] [PubMed] [Google Scholar]
- 118.Tsubuki S, Takaki Y & Saido TC Dutch, Flemish, Italian, and Arctic mutations of APP and resistance of Abeta to physiologically relevant proteolytic degradation. Lancet 361, 1957–1958 (2003). [DOI] [PubMed] [Google Scholar]
- 119.Morelli L et al. Differential degradation of amyloid beta genetic variants associated with hereditary dementia or stroke by insulin-degrading enzyme. J. Biol. Chem 278, 23221–23226 (2003). [DOI] [PubMed] [Google Scholar]
- 120.Monro OR et al. Substitution at codon 22 reduces clearance of Alzheimer's amyloid-beta peptide from the cerebrospinal fluid and prevents its transport from the central nervous system into blood. Neurobiol. Aging 23, 405–412 (2002). [DOI] [PubMed] [Google Scholar]
- 121.Sleegers K et al. APP duplication is sufficient to cause early onset Alzheimer's dementia with cerebral amyloid angiopathy. Brain 129, 2977–2983 (2006). [DOI] [PubMed] [Google Scholar]
- 122.Rovelet-Lecrux A et al. APP locus duplication causes autosomal dominant early-onset Alzheimer disease with cerebral amyloid angiopathy. Nat. Genet 38, 24–26 (2006). [DOI] [PubMed] [Google Scholar]
- 123.Wilcock DM, Schmitt FA & Head E Cerebrovascular contributions to aging and Alzheimer's disease in Down syndrome. Biochim Biophys Acta 1862, 909–914 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Nochlin D, Bird TD, Nemens EJ, Ball MJ & Sumi SM Amyloid angiopathy in a Volga German family with Alzheimer's disease and a presenilin-2 mutation (N141I). Ann. Neurol 43, 131–135 (1998). [DOI] [PubMed] [Google Scholar]
- 125.Dermaut B et al. Cerebral amyloid angiopathy is a pathogenic lesion in Alzheimer's disease due to a novel presenilin 1 mutation. Brain 124, 2383–2392 (2001). [DOI] [PubMed] [Google Scholar]
- 126.Sanchez-Valle R et al. A novel mutation in the PSEN1 gene (L286P) associated with familial early-onset dementia of Alzheimer type and lobar haematomas. Eur J Neurol 14, 1409–1412 (2007). [DOI] [PubMed] [Google Scholar]
- 127.Mann DM, Pickering-Brown SM, Takeuchi A & Iwatsubo T Amyloid angiopathy and variability in amyloid beta deposition is determined by mutation position in presenilin-1-linked Alzheimer's disease. The American journal of pathology 158, 2165–2175 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Ryan NS et al. Genetic determinants of white matter hyperintensities and amyloid angiopathy in familial Alzheimer's disease. Neurobiol. Aging 36, 3140–3151 (2015). [DOI] [PubMed] [Google Scholar]
- 129.Woo D et al. Meta-analysis of genome-wide association studies identifies 1q22 as a susceptibility locus for intracerebral hemorrhage. Am. J. Hum. Genet 94, 511–521 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Marini S et al. Association of Apolipoprotein E With Intracerebral Hemorrhage Risk by Race/Ethnicity: A Meta-analysis. JAMA Neurol. 76, 480–491 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Beecham GW et al. Genome-wide association meta-analysis of neuropathologic features of Alzheimer's disease and related dementias. PLoS Genet 10, e1004606 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]; In this study and reference 132, associations between CAA pathology and genetic variants that promote Alzheimer disease pathology were examined.
- 132.Makela M et al. Alzheimer risk loci and associated neuropathology in a population-based study (Vantaa 85+). Neurol Genet 4, e211 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Eikelenboom P & Stam FC Immunoglobulins and complement factors in senile plaques. An immunoperoxidase study. Acta Neuropathol. 57, 239–242 (1982). [DOI] [PubMed] [Google Scholar]
- 134.Snow AD et al. The presence of heparan sulfate proteoglycans in the neuritic plaques and congophilic angiopathy in Alzheimer's disease. Am. J. Pathol 133, 456–463 (1988). [PMC free article] [PubMed] [Google Scholar]
- 135.Kalaria RN & Grahovac I Serum amyloid P immunoreactivity in hippocampal tangles, plaques and vessels: implications for leakage across the blood-brain barrier in Alzheimer's disease. Brain Res. 516, 349–353 (1990). [DOI] [PubMed] [Google Scholar]
- 136.Rozemuller JM et al. Distribution pattern and functional state of alpha 1-antichymotrypsin in plaques and vascular amyloid in Alzheimer's disease. A immunohistochemical study with monoclonal antibodies against native and inactivated alpha 1-antichymotrypsin. Acta Neuropathol. 82, 200–207 (1991). [DOI] [PubMed] [Google Scholar]
- 137.Van Gool D, De Strooper B, Van Leuven F, Triau E & Dom R alpha 2-Macroglobulin expression in neuritic-type plaques in patients with Alzheimer's disease. Neurobiol. Aging 14, 233–237 (1993). [DOI] [PubMed] [Google Scholar]
- 138.Verbeek MM et al. Accumulation of intercellular adhesion molecule-1 in senile plaques in brain tissue of patients with Alzheimer's disease. Am. J. Pathol 144, 104–116 (1994). [PMC free article] [PubMed] [Google Scholar]
- 139.McGeer PL, Klegeris A, Walker DG, Yasuhara O & McGeer EG Pathological proteins in senile plaques. Tohoku J Exp Med 174, 269–277 (1994). [DOI] [PubMed] [Google Scholar]
- 140.Harr SD, Uint L, Hollister R, Hyman BT & Mendez AJ Brain expression of apolipoproteins E, J, and A-I in Alzheimer's disease. J. Neurochem 66, 2429–2435 (1996). [DOI] [PubMed] [Google Scholar]
- 141.Verbeek MM, Otte-Holler I, Veerhuis R, Ruiter DJ & De Waal RM Distribution of A beta-associated proteins in cerebrovascular amyloid of Alzheimer's disease. Acta Neuropathol (Berl) 96, 628–636 (1998). [DOI] [PubMed] [Google Scholar]
- 142.Hashimoto T et al. CLAC: a novel Alzheimer amyloid plaque component derived from a transmembrane precursor, CLAC-P/collagen type XXV. EMBO J. 21, 1524–1534 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Kowa H et al. Mostly separate distributions of CLAC-versus Abeta40- or thioflavin S-reactivities in senile plaques reveal two distinct subpopulations of beta-amyloid deposits. Am. J. Pathol 165, 273–281 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Zabel M et al. A shift in microglial beta-amyloid binding in Alzheimer's disease is associated with cerebral amyloid angiopathy. Brain Pathol 23, 390–401 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Camacho J et al. Brain ApoA-I, ApoJ and ApoE Immunodetection in Cerebral Amyloid Angiopathy. Front Neurol 10, 187 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Ghiso J et al. The cerebrospinal-fluid soluble form of Alzheimer's amyloid beta is complexed to SP-40,40 (apolipoprotein J), an inhibitor of the complement membrane-attack complex. Biochem J 293, 27–30 (1993). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Webster S, O'Barr S & Rogers J Enhanced aggregation and beta structure of amyloid beta peptide after coincubation with C1q. J. Neurosci. Res 39, 448–456 (1994). [DOI] [PubMed] [Google Scholar]
- 148.Snow AD et al. An important role of heparan sulfate proteoglycan (Perlecan) in a model system for the deposition and persistence of fibrillar A beta-amyloid in rat brain. Neuron 12, 219–234 (1994). [DOI] [PubMed] [Google Scholar]
- 149.Ma J, Yee A, Brewer HBJ, Das S & Potter H Amyloid-associated proteins alpha 1-antichymotrypsin and apolipoprotein E promote assembly of Alzheimer beta-protein into filaments. Nature 372, 92–94 (1994). [DOI] [PubMed] [Google Scholar]
- 150.Endo Y et al. Apolipoprotein E and clusterin inhibit the early phase of amyloid-beta aggregation in an in vitro model of cerebral amyloid angiopathy. Acta Neuropathol Commun 7, 12 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Namba Y, Tomonaga M, Kawasaki H, Otomo E & Ikeda K Apolipoprotein E immunoreactivity in cerebral amyloid deposits and neurofibrillary tangles in Alzheimer's disease and kuru plaque amyloid in Creutzfeldt-Jakob disease. Brain Res. 541, 163–166 (1991). [DOI] [PubMed] [Google Scholar]
- 152.Wisniewski T, Golabek AA, Kida E, Wisniewski KE & Frangione B Conformational mimicry in Alzheimer's disease. Role of apolipoproteins in amyloidogenesis. Am. J. Pathol 147, 238–244 (1995). [PMC free article] [PubMed] [Google Scholar]
- 153.Strittmatter WJ et al. Apolipoprotein E: high-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc. Natl. Acad. Sci. USA 90, 1977–1981 (1993). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Liu Y et al. APOE genotype and neuroimaging markers of Alzheimer's disease: systematic review and meta-analysis. J Neurol Neurosurg Psychiatry 86, 127–134 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Greenberg SM, Rebeck GW, Vonsattel JPV, Gomez-Isla T & Hyman BT Apolipoprotein E e4 and cerebral hemorrhage associated with amyloid angiopathy. Ann. Neurol 38, 254–259 (1995). [DOI] [PubMed] [Google Scholar]
- 156.Nicoll JA & McCarron MO APOE gene polymorphism as a risk factor for cerebral amyloid angiopathy-related hemorrhage. Amyloid 8 (Suppl. 1), 51–55 (2001). [PubMed] [Google Scholar]
- 157.Olichney JM et al. The apolipoprotein E epsilon 4 allele is associated with increased neuritic plaques and cerebral amyloid angiopathy in Alzheimer's disease and Lewy body variant. Neurology 47, 190–196 (1996). [DOI] [PubMed] [Google Scholar]
- 158.Corder EH et al. Protective effect of apolipoprotein E type 2 allele for late onset Alzheimer disease. Nat. Genet 7, 180–184 (1994). [DOI] [PubMed] [Google Scholar]
- 159.Deane R et al. apoE isoform-specific disruption of amyloid beta peptide clearance from mouse brain. J. Clin. Invest 118, 4002–4013 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Verghese PB et al. ApoE influences amyloid-beta (Abeta) clearance despite minimal apoE/Abeta association in physiological conditions. Proc Natl Acad Sci U S A 110, E1807–1816 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Huynh TV, Davis AA, Ulrich JD & Holtzman DM Apolipoprotein E and Alzheimer's disease: the influence of apolipoprotein E on amyloid-beta and other amyloidogenic proteins. J Lipid Res 58, 824–836 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Guo S et al. Effects of apoE isoforms on beta-amyloid-induced matrix metalloproteinase-9 in rat astrocytes. Brain Res. 1111, 222–226 (2006). [DOI] [PubMed] [Google Scholar]
- 163.Bales KR et al. Lack of apolipoprotein E dramatically reduces amyloid beta-peptide deposition. Nat. Genet 17, 263–264 (1997). [DOI] [PubMed] [Google Scholar]
- 164.Fryer JD et al. Apolipoprotein E markedly facilitates age-dependent cerebral amyloid angiopathy and spontaneous hemorrhage in amyloid precursor protein transgenic mice. J. Neurosci 23, 7889–7896 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study and references 165–167 demonstrate the complex relationship between CAA pathology and the presence of apolipoprotein E, its isoforms, and its human and mouse forms.
- 165.Holtzman DM et al. Apolipoprotein E isoform-dependent amyloid deposition and neuritic degeneration in a mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A 97, 2892–2897. (2000). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Holtzman DM et al. Expression of human apolipoprotein E reduces amyloid-beta deposition in a mouse model of Alzheimer's disease. J. Clin. Invest 103, 15–21 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Fryer JD et al. Human apolipoprotein E4 alters the amyloid-beta 40:42 ratio and promotes the formation of cerebral amyloid angiopathy in an amyloid precursor protein transgenic model. J. Neurosci 25, 2803–2810 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Liao F et al. Murine versus human apolipoprotein E4: differential facilitation of and colocalization in cerebral amyloid angiopathy and amyloid plaques in APP transgenic mouse models. Acta Neuropathol Commun 3, 70 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Thal DR et al. Capillary cerebral amyloid angiopathy identifies a distinct APOE epsilon4-associated subtype of sporadic Alzheimer's disease. Acta Neuropathol. 120, 169–183 (2010). [DOI] [PubMed] [Google Scholar]
- 170.McGeer PL, Kawamata T & Walker DG Distribution of clusterin in Alzheimer brain tissue. Brain Res. 579, 337–341 (1992). [DOI] [PubMed] [Google Scholar]
- 171.Miners JS, Clarke P & Love S Clusterin levels are increased in Alzheimer's disease and influence the regional distribution of Abeta. Brain Pathol 27, 305–313 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Hammad SM, Ranganathan S, Loukinova E, Twal WO & Argraves WS Interaction of apolipoprotein J-amyloid beta-peptide complex with low density lipoprotein receptor-related protein-2/megalin. A mechanism to prevent pathological accumulation of amyloid beta-peptide. J. Biol. Chem 272, 18644–18649 (1997). [DOI] [PubMed] [Google Scholar]
- 173.Narayan P et al. The extracellular chaperone clusterin sequesters oligomeric forms of the amyloid-beta(1–40) peptide. Nat Struct Mol Biol 19, 79–83 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Yerbury JJ et al. The extracellular chaperone clusterin influences amyloid formation and toxicity by interacting with prefibrillar structures. FASEB J. 21, 2312–2322 (2007). [DOI] [PubMed] [Google Scholar]
- 175.Wojtas AM et al. Loss of clusterin shifts amyloid deposition to the cerebrovasculature via disruption of perivascular drainage pathways. Proc Natl Acad Sci U S A 114, E6962–E6971 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Bell RD et al. Transport pathways for clearance of human Alzheimer's amyloid beta-peptide and apolipoproteins E and J in the mouse central nervous system. J Cereb Blood Flow Metab 27, 909–918 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.DeMattos RB et al. Clusterin promotes amyloid plaque formation and is critical for neuritic toxicity in a mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A 99, 10843–10848 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Oh SB et al. Clusterin contributes to early stage of Alzheimer's disease pathogenesis. Brain Pathol 29, 217–231 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Drummond E et al. Proteomic differences in amyloid plaques in rapidly progressive and sporadic Alzheimer's disease. Acta Neuropathol. 133, 933–954 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Xiong F, Ge W & Ma C Quantitative proteomics reveals distinct composition of amyloid plaques in Alzheimer's disease. Alzheimers Dement 15, 429–440 (2019). [DOI] [PubMed] [Google Scholar]
- 181.Manousopoulou A et al. Systems proteomic analysis reveals that clusterin and tissue inhibitor of metalloproteinases 3 increase in leptomeningeal arteries affected by cerebral amyloid angiopathy. Neuropathol Appl Neurobiol 43, 492–504 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Hondius DC et al. Proteomics analysis identifies new markers associated with capillary cerebral amyloid angiopathy in Alzheimer's disease. Acta Neuropathol Commun 6, 46 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Inoue Y et al. Sushi repeat-containing protein 1: a novel disease-associated molecule in cerebral amyloid angiopathy. Acta Neuropathol. 134, 605–617 (2017). [DOI] [PubMed] [Google Scholar]
- 184.Cortes-Canteli M et al. Fibrinogen and beta-amyloid association alters thrombosis and fibrinolysis: a possible contributing factor to Alzheimer's disease. Neuron 66, 695–709 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Hultman K, Strickland S & Norris EH The APOE varepsilon4/varepsilon4 genotype potentiates vascular fibrin(ogen) deposition in amyloid-laden vessels in the brains of Alzheimer's disease patients. J Cereb Blood Flow Metab 33, 1251–1258 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Sperling RA et al. Amyloid-related imaging abnormalities in amyloid-modifying therapeutic trials: recommendations from the Alzheimer's Association Research Roundtable Workgroup. Alzheimers Dement 7, 367–385 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Salloway S et al. Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer's disease. The New England journal of medicine 370, 322–333 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Ostrowitzki S et al. A phase III randomized trial of gantenerumab in prodromal Alzheimer's disease. Alzheimers Res Ther 9, 95 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Sevigny J et al. The antibody aducanumab reduces Abeta plaques in Alzheimer's disease. Nature 537, 50–56 (2016). [DOI] [PubMed] [Google Scholar]
- 190.Carlson C et al. Amyloid-related imaging abnormalities from trials of solanezumab for Alzheimer's disease. Alzheimers Dement (Amst) 2, 75–85 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Cummings JL et al. ABBY: A phase 2 randomized trial of crenezumab in mild to moderate Alzheimer disease. Neurology 90, e1889–e1897 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Leurent C et al. Immunotherapy with ponezumab for probable cerebral amyloid angiopathy. Ann Clin Transl Neurol (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]; The initial clinic trial of anti-amyloid-β immunotherapy for patients diagnosed with CAA cerebral amyloid angiopathy.
- 193.Ultsch M et al. Structure of Crenezumab Complex with Abeta Shows Loss of beta-Hairpin. Sci Rep 6, 39374 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Raman MR et al. Spontaneous amyloid-related imaging abnormalities in a cognitively normal adult. Neurology 83, 1771–1772 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Ryan NS et al. Spontaneous ARIA (amyloid-related imaging abnormalities) and cerebral amyloid angiopathy related inflammation in presenilin 1-associated familial Alzheimer's disease. J Alzheimers Dis 44, 1069–1074 (2015). [DOI] [PubMed] [Google Scholar]
- 196.Ketter N et al. Central Review of Amyloid-Related Imaging Abnormalities in Two Phase III Clinical Trials of Bapineuzumab in Mild-To-Moderate Alzheimer's Disease Patients. J Alzheimers Dis 57, 557–573 (2017). [DOI] [PubMed] [Google Scholar]
- 197.Penninkilampi R, Brothers HM & Eslick GD Safety and Efficacy of Anti-Amyloid-beta Immunotherapy in Alzheimer's Disease: A Systematic Review and Meta-Analysis. J Neuroimmune Pharmacol 12, 194–203 (2017). [DOI] [PubMed] [Google Scholar]
- 198.Goos JD et al. Incidence of cerebral microbleeds: a longitudinal study in a memory clinic population. Neurology 74, 1954–1960 (2010). [DOI] [PubMed] [Google Scholar]
- 199.Haeberlein SB et al. 24-Month analysis of change from baseline in clinical dementia rating scale cognitive and functional domains in PRIME: A randomized phase 1B study of the anti-amyloid beta monoclonal antibody aducanumab. Alzheimer’s & dementia : the journal of the Alzheimer's Association 14, P242 (2018). [Google Scholar]
- 200.Andjelkovic M et al. Update on the safety and tolerability of gangenerumab in the ongoing open-label extension of the SCarlet RoAD study in patients with prodromal Alzheimer’s disease after approximately 2 years of study duration. Alzheimer’s & dementia : the journal of the Alzheimer's Association 14, P241–P242 (2018). [Google Scholar]
- 201.Sperling R et al. Amyloid-related imaging abnormalities in patients with Alzheimer's disease treated with bapineuzumab: a retrospective analysis. Lancet Neurol 11, 241–249 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]; An analysis of amyloid-related imaging abnormalities in clinical trials of the anti-amyloid-β antibody bapineuzumab and in which candidate mechanisms were proposed.
- 202.Arrighi HM et al. Amyloid-related imaging abnormalities-haemosiderin (ARIA-H) in patients with Alzheimer's disease treated with bapineuzumab: a historical, prospective secondary analysis. J Neurol Neurosurg Psychiatry 87, 106–112 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Brashear HR et al. Clinical Evaluation of Amyloid-Related Imaging Abnormalities in Bapineuzumab Phase III Studies. J Alzheimers Dis 66, 1409–1424 (2018). [DOI] [PubMed] [Google Scholar]
- 204.Liu E et al. Biomarker pattern of ARIA-E participants in phase 3 randomized clinical trials with bapineuzumab. Neurology 90, e877–e886 (2018). [DOI] [PubMed] [Google Scholar]
- 205.Boche D, Denham N, Holmes C & Nicoll JA Neuropathology after active Abeta42 immunotherapy: implications for Alzheimer's disease pathogenesis. Acta Neuropathol. 120, 369–384 (2010). [DOI] [PubMed] [Google Scholar]
- 206.Sakai K et al. Abeta immunotherapy for Alzheimer's disease: effects on apoE and cerebral vasculopathy. Acta Neuropathol. 128, 777–789 (2014). [DOI] [PubMed] [Google Scholar]
- 207.Mandybur TI Cerebral amyloid angiopathy: the vascular pathology and complications. Journal of neuropathology and experimental neurology 45, 79–90 (1986). [PubMed] [Google Scholar]
- 208.Kinnecom C et al. Course of cerebral amyloid angiopathy-related inflammation. Neurology 68, 1411–1416 (2007). [DOI] [PubMed] [Google Scholar]
- 209.Auriel E et al. Validation of Clinicoradiological Criteria for the Diagnosis of Cerebral Amyloid Angiopathy-Related Inflammation. JAMA Neurol 73, 197–202 (2016). [DOI] [PubMed] [Google Scholar]
- 210.Piazza F et al. Anti-amyloid beta autoantibodies in cerebral amyloid angiopathy-related inflammation: Implications for Amyloid-Modifying Therapies. Ann. Neurol 73, 449–458 (2013). [DOI] [PubMed] [Google Scholar]; The first report of anti-amyloid-β antibodies in the CSF of individuals with CAA, offering the strongest link between this spontaneous syndrome and ARIA seen in clinical trials of anti-amyloid-β antibodies.
- 211.Zago W et al. Vascular alterations in PDAPP mice after anti-Abeta immunotherapy: Implications for amyloid-related imaging abnormalities. Alzheimers Dement (2013). [DOI] [PubMed] [Google Scholar]
- 212.Blockx I et al. Monitoring Blood-Brain Barrier Integrity Following Amyloid-beta Immunotherapy Using Gadolinium-Enhanced MRI in a PDAPP Mouse Model. J Alzheimers Dis 54, 723–735 (2016). [DOI] [PubMed] [Google Scholar]
- 213.Gottesman RF et al. Associations Between Midlife Vascular Risk Factors and 25-Year Incident Dementia in the Atherosclerosis Risk in Communities (ARIC) Cohort. JAMA Neurol 74, 1246–1254 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Boche D et al. Consequence of Abeta immunization on the vasculature of human Alzheimer's disease brain. Brain 131, 3299–3310 (2008). [DOI] [PubMed] [Google Scholar]; This study identified changes in CAA severity in post-mortem brain tissue from individiuals who were previously immunized against amyloid-β.
- 215.Han BH et al. Resorufin analogs preferentially bind cerebrovascular amyloid: potential use as imaging ligands for cerebral amyloid angiopathy. Molecular neurodegeneration 6, 86 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Mawuenyega KG et al. Decreased clearance of CNS beta-amyloid in Alzheimer's disease. Science 330, 1774 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.de Leon MJ et al. Cerebrospinal Fluid Clearance in Alzheimer Disease Measured with Dynamic PET. J Nucl Med 58, 1471–1476 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Goate A et al. Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer's disease. Nature 349, 704–706 (1991). [DOI] [PubMed] [Google Scholar]
- 219.Mullan M et al. A pathogenic mutation for probable Alzheimer's disease in the APP gene at the N-terminus of beta-amyloid. Nat. Genet 1, 345–347 (1992). [DOI] [PubMed] [Google Scholar]
- 220.Rovelet-Lecrux A et al. APP locus duplication in a Finnish family with dementia and intracerebral haemorrhage. J Neurol Neurosurg Psychiatry 78, 1158–1159 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Belza MG & Urich H Cerebral amyloid angiopathy in Down's syndrome. Clinical neuropathology 5, 257–260 (1986). [PubMed] [Google Scholar]
- 222.Nicoll JA et al. High frequency of apolipoprotein E epsilon 2 allele in hemorrhage due to cerebral amyloid angiopathy. Ann. Neurol 41, 716–721 (1997). [DOI] [PubMed] [Google Scholar]
- 223.Greenberg SM et al. Association of apolipoprotein E epsilon2 and vasculopathy in cerebral amyloid angiopathy. Neurology 50, 961–965 (1998). [DOI] [PubMed] [Google Scholar]
- 224.Cotman SL, Halfter W & Cole GJ Agrin binds to beta-amyloid (Abeta), accelerates abeta fibril formation, and is localized to Abeta deposits in Alzheimer's disease brain. Mol Cell Neurosci 15, 183–198 (2000). [DOI] [PubMed] [Google Scholar]
- 225.van Horssen J et al. Heparan sulfate proteoglycan expression in cerebrovascular amyloid beta deposits in Alzheimer's disease and hereditary cerebral hemorrhage with amyloidosis (Dutch) brains. Acta Neuropathol. 102, 604–614 (2001). [DOI] [PubMed] [Google Scholar]
- 226.Wilhelmus MM et al. Specific association of small heat shock proteins with the pathological hallmarks of Alzheimer's disease brains. Neuropathol Appl Neurobiol 32, 119–130 (2006). [DOI] [PubMed] [Google Scholar]
- 227.Wilhelmus MM et al. Small heat shock protein HspB8: its distribution in Alzheimer's disease brains and its inhibition of amyloid-beta protein aggregation and cerebrovascular amyloid-beta toxicity. Acta Neuropathol. 111, 139–149 (2006). [DOI] [PubMed] [Google Scholar]
- 228.van Horssen J et al. Collagen XVIII: a novel heparan sulfate proteoglycan associated with vascular amyloid depositions and senile plaques in Alzheimer's disease brains. Brain Pathol 12, 456–462 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Verbeek MM, Otte HI, Wesseling P, Ruiter DJ & de WR Induction of alpha-smooth muscle actin expression in cultured human brain pericytes by transforming growth factor-beta 1. Am. J. Pathol 144, 372–382 (1994). [PMC free article] [PubMed] [Google Scholar]
- 230.Zhao L et al. Matrix metalloproteinase 9-mediated intracerebral hemorrhage induced by cerebral amyloid angiopathy. Neurobiol. Aging 36, 2963–2971 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]