Abstract
Cerebrovascular disease underpins vascular dementia (VaD), but structural and functional changes to the cerebral vasculature contribute to disease pathology and cognitive decline in Alzheimer's disease (AD). In this review, we discuss the contribution of cerebral amyloid angiopathy and non‐amyloid small vessel disease in AD, and the accompanying changes to the density, maintenance and remodelling of vessels (including alterations to the composition and function of the cerebrovascular basement membrane). We consider how abnormalities of the constituent cells of the neurovascular unit – particularly of endothelial cells and pericytes – and impairment of the blood‐brain barrier (BBB) impact on the pathogenesis of AD. We also discuss how changes to the cerebral vasculature are likely to impair Aβ clearance – both intra‐periarteriolar drainage (IPAD) and transport of Aβ peptides across the BBB, and how impaired neurovascular coupling and reduced blood flow in relation to metabolic demand increase amyloidogenic processing of APP and the production of Aβ. We review the vasoactive properties of Aβ peptides themselves, and the probable bi‐directional relationship between vascular dysfunction and Aβ accumulation in AD. Lastly, we discuss recent methodological advances in transcriptomics and imaging that have provided novel insights into vascular changes in AD, and recent advances in assessment of the retina that allow in vivo detection of vascular changes in the early stages of AD.
Keywords: Alzheimer's disease, blood‐brain barrier, cerebral blood flow, neurovascular coupling, pericyte, vasculature
Structural and functional changes to blood vessels not only underpin disease pathogenesis in vascular dementia but are also major contributors to cognitive decline and disease progression in Alzheimer’s disease. We provide an updated review on the clinical and neuropathological vascular features (SVD, CAA) and underpinning functional changes to blood vessels in relation of disease pathology in Alzheimer’s disease. We also discuss recent methodological advances within the field that have provided novel insights into vascular changes in AD, including transcriptomics, imaging, and retinal assessment providing a window of opportunity for future clinical intervention.
1. CEREBROVASCULAR DISEASE AND ALZHEIMER'S DISEASE
Alzheimer's disease (AD) and vascular dementia (VaD) account for approximately 60%–80% and 5%–10% of patients with dementia, based on clinical [1] and neuropathologic diagnosis [2]. Most patients, including the majority with AD, have mixed pathologies that include pathological evidence of cerebrovascular disease [3, 4, 5]. Ischaemic damage to the white matter, attributed to small vessel disease (SVD), is associated with an increased risk of developing AD [6], and over 90% of AD patients have cerebral amyloid angiopathy (CAA; Figure 1E) [7, 8, 9]. In VaD, cerebral ischaemia is the defining pathological process, usually secondary to non‐amyloid, arteriolosclerotic small vessel disease (NA‐SVD; Figure 1A‐B) [10, 11], though also linked to ischaemic stroke injury [12]. There is also evidence of NA‐SVD comorbidities in AD cases [13, 14, 15] and the presence of NA‐SVD may be a risk factor for the development of AD [16].
FIGURE 1.
Common vasculopathies in AD. (A–D) Haematoxylin and eosin staining of fixed brain tissue sections with (A) normal white matter arterioles; (B) arterioles and artery with severe sclerosis; (C) artery with microatheroma, (D); non‐amyloid SVD‐associated microhaemorrhages – old (arrows) and more recent (double arrow) and a microinfarct (arrowheads). (E) Immunoperoxidase labelling of Aβ revealing Type‐2 CAA. (F) Immunofluorescent labelling of collagen (green) with DAPI (blue) nuclear stain reveals a string vessel (white arrow) connecting adjacent capillaries. Scale bars: (A) 100 µm (B) 100 µm (C) 200 µm (D) 100 µm (E) 50 µm (F) 50 µm.
A large scale multifactorial analysis of brain images from the Alzheimer's Disease Neuroimaging Initiative (ADNI) and modelling of late‐onset AD (LOAD) suggest that vascular dysregulation is an early and possibly an initial pathological event in AD [17, 18]. Cerebral hypoperfusion [19, 20] and blood‐brain barrier (BBB) breakdown [21, 22] precede the clinical presentation of dementia. Though BBB breakdown does occur in normal ageing, it is exacerbated in the early stages of AD, particularly within the hippocampus [21], and is associated with cognitive decline independently of changes in Aβ and Tau [23, 24]. In familial AD, reduction in cerebral blood flow (CBF) and glucose uptake occur soon after initial Aβ deposition and well before clinical disease [25, 26, 27]. There is also increasing evidence that malfunction of the neurovascular unit, partly related to injury to pericytes, is an early contributor to the development of AD [21, 28, 29].
Pathological changes to the cerebral vasculature influence several processes involved in the progression of AD. Damage to the vasculature impairs periarteriolar clearance of Aβ (reviewed here Refs. [30, 31, 32, 33]) and receptor‐mediated removal of Aβ peptides across the BBB (reviewed here Refs. [34, 35, 36, 37]), accelerating Aβ deposition. Reduced cerebral oxygenation as a result of diminished blood flow and impaired neurovascular coupling enhance amyloidogenic processing of APP (Figure 2; reviews of in vitro and in vivo evidence here [5, 38]). Aβ peptides themselves are vasoactive, inducing contraction of pericytes [39] and vascular smooth muscle cells [40] which exacerbates hypoperfusion [39], and impairs BBB function [41]. Strong relationships have also been reported between measures of vascular dysfunction and the accumulation of phospho‐tau [42, 43, 44, 45] and TDP‐43 [11, 46] indicating that vascular dysfunction extends beyond Aβ pathology. Several therapeutic interventions have been proposed for the prevention or treatment of AD through improving cerebral hypoperfusion; these include the administration of vascular growth factors (reviewed here Ref. [47]).
FIGURE 2.
Vascular dysfunction and Aβ homeostasis. Aβ transport. Depletion of receptors involved in the transport of Aβ out of the brain parenchyma such as LRP‐1, upregulation of RAGE which transports Aβ into the parenchyma, impaired perivascular drainage, reduction of Aβ degradation, and BBB breakdown all contribute to Aβ accumulation in the brain in AD. Increased Aβ exacerbates BBB breakdown and CVBM alterations, constituting a vicious pathological cycle. Blood flow. Ischaemia elevates BACE‐1 activity which increases the processing of APP to Aβ, leading to oxidative stress, and upregulation of the vasoconstrictors angiotensin‐II (Ang‐II) and endothelin‐1 (EDN1). These stimulate pericyte constriction, lowering blood flow and exacerbating ischaemia. Similar pathological cycles may exist not only for Aβ but for other proteins associated with AD such as tau and TDP‐43
Here we review the literature and provide an update on the structural, morphological, and functional changes to the cerebral vasculature in AD and discuss novel methods for investigating the cerebral vasculature in AD including examination of the retina.
2. COMMON VASCULAR PATHOLOGIES IN AD: CEREBRAL AMYLOID ANGIOPATHY, AND NON‐AMYLOID SMALL VESSEL DISEASE
Cerebral small vessel disease (SVD) is a major contributor to cognitive impairment in VaD (including post‐stroke dementia), and a frequent co‐pathology in AD [6, 10, 48, 49, 50, 51, 52, 53]. Neuroimaging features of SVD include white matter hyperintensities, infarcts, lacunae, haemorrhage, microbleeds and enlarged perivascular spaces – commonly reported in VaD and AD [52, 54, 55, 56, 57]. There are several types of SVD, the two most common being cerebral amyloid angiopathy (CAA) and non‐amyloid SVD (NA‐SVD) [51, 52].
2.1. Cerebral amyloid angiopathy
Aβ deposits not only in the brain parenchyma as plaques, but also in the walls of blood vessels, particularly arterioles, causing CAA. CAA affects 30%–40% of elderly people without dementia, the proportion increasing with age from about 65 years [58, 59], and over 80% of people with AD [7, 8, 9, 60, 61 ]. CAA is associated with increased risk of dementia, and more rapid cognitive decline in AD [62]. The severity of CAA has been assessed using a validated semiquantitative protocol [63] or by counting the number of neuroanatomical regions affected by CAA [64]. Arteriolar CAA and capCAA are most prevalent in the occipital lobe (>92% and 35%–47% of AD cases; 86% and 21% of non‐AD controls [63]).
APOE influences the risk of developing sporadic CAA [56, 65]. The severity of arteriolar CAA is increased by possession of APOE ε4 [7, 8, 9, 66, 67] which is also strongly associated with capillary CAA [63, 68, 69]. CAA is less prevalent in APOE ε2 suggestive of protection [63, 68, 69].
Sporadic CAA has been subdivided into two types. CAA‐Type 1 is more commonly associated with APOE ε4 and is characterised by Aβ deposition in cortical capillaries (capCAA) in addition to larger cortical and leptomeningeal vessels. In CAA‐Type 2, associated less strongly with APOE ε4 but also with ε2, Aβ accumulates in arteries, arterioles, veins and venules but not capillaries [63, 68, 70, 71] (Figure 1E). Parenchymal plaques consist of Aβ isoforms that tend to terminate at the 42nd amino acid, often modified with pyroglutamate [72]. Arteriolar deposits in CAA are mostly shorter Aβ1‐40 and Aβ2‐40 [72, 73, 74, 75, 76], though Aβ1‐42 predominates in capillary CAA [72, 77, 78]. Vascular Aβ peptides are likely to be of neuronal origin, and their deposition in the walls of blood vessels is probably promoted by impaired perivascular clearance (see below).
CAA can be familial or sporadic. Familial CAA is rare but often more severe, manifesting clinically at an earlier age than sporadic CAA [79]. Familial Aβ‐CAA is associated with APP mutations or duplications, or mutations in PSEN1 or PSEN2 [56]; several APP‐linked mutations are used in transgenic animals to model CAA (reviewed here Refs. [80, 81]). Rarer forms of familial CAA are caused by vascular deposition of other amyloid proteins, including cystatin C, transthyretin, gelsolin, prion protein and BRI2‐gene products [82].
Arteriolar deposition of Aβ commences in the extracellular matrix of the tunica media but may progress to replace all SMCs and other tissue elements within the vessel wall [83]. Several additional vasculopathic abnormalities may complicate CAA, including fibrinoid necrosis, microaneurysms, concentric splitting of the vessel wall, and hyaline vessel wall thickening and arteriolar degeneration, and formation of glomeruloid capillary clusters [55, 63, 84, 85]. These vasculopathies likely underlie the cerebrovascular dysfunction found in CAA, several of which have been directly associated with microbleeds and haemorrhage [84, 85, 86, 87, 88]. APOE ε2 is associated with both fibrinoid necrosis and CAA‐associated haemorrhage [89]. Another rarer but increasingly recognised vasculopathy is CAA‐related inflammation (CAA‐RI), which can be subtyped into inflammatory CAA (ICAA) and Aβ‐related angiitis (ABRA), often treatable with immunosuppressive therapy (reviewed here Refs. [90, 91]). Clinicoradiological criteria allow diagnosis of probable CAA‐RI, avoiding the need for biopsy in most cases [92].
A recent in vivo imaging study of APP/PS1 mice with CAA found microhaemorrhages mostly at vessel bifurcations or bends without amyloid deposition [93]. The authors posited that altered flow dynamics within CAA‐affected vessels cause blood leakage. In capCAA, Aβ accumulation in the vascular basement membrane leads to degeneration of endothelial cells, loss of TJs and BBB breakdown [94], often associated with severe AD pathology [71, 78]. As sometimes occurs in severe “dyshoric” arteriolar CAA [9], Aβ deposits in capCAA tend to extend into the adjacent brain parenchyma where they are associated with neuroinflammation and hyperphosphorylated tau [68, 95, 96].
CAA increases the risk of cerebral haemorrhage and infarction [9, 56]. Common clinical MRI imaging features include lobar intracerebral haemorrhage, microhaemorrhages, siderosis, and white matter hyperintensities (WMHs) [56, 88, 97, 98, 99 ]. CAA progressively decreases vascular reactivity [100, 101, 102, 103], increasing the probability of hypoperfusion and ischaemic brain damage. CAA is associated with cortical atrophy independent of AD [104]. Functional brain connectivity was shown to be attenuated in Dutch type hereditary CAA [105].
Sporadic CAA probably results from impaired clearance of perivascular Aβ from the brain [106, 107]. Diminished intra‐mural periarterial drainage (IPAD) and perivascular CSF influx [6, 83, 106, 108, 109, 110], endothelial transport across the BBB [68, 70, 111, 112], or enzymatic degradation of Aβ [113, 114] may all contribute. By impairing vasomotion, Aβ‐mediated dysfunction of vascular smooth muscle may impede intramural clearance of Aβ within the interstitial fluid [115].
2.2. Non‐amyloid small vessel disease
NA‐SVD, sometimes referred to as ‘hypertensive angiopathy’, typically affects the small perforating arteries of the deep grey and white matter [51, 52]. Half of VaD cases are preceded by mild vascular cognitive impairment, which is also associated with NA‐SVD [15, 116, 117]. There is debate as to whether NA‐SVD may increase the likelihood of developing AD [16]. Age, smoking, diabetes and hypertension are risk factors for NA‐SVD [5, 15, 51, 52]. The genetic risk factors for NA‐SVD are still unclear but variation at several gene loci has been associated with its typical clinical and radiological manifestations [118]. We proposed previously that hypertension could be not only a cause but also a consequence of NA‐SVD: a cardiovascular response that is induced to maintain cerebral perfusion in the face of increasing cerebral vascular resistance [15]. This presents the possibility of a vicious cycle between hypertension and NA‐SVD, a possibility supported by our recent demonstration that although late‐life hypertension was associated with markers of vascular damage (SVD severity and BBB breakdown), it was also associated with evidence of better cerebral perfusion and lower insoluble Aβ42 levels in AD and mixed dementia [119].
The core pathology of NA‐SVD is arteriolosclerosis (Figure 1B): collagenous thickening of the vessel wall, narrowing of the vessel lumen and loss of SMCs. NA‐SVD can be further categorised by the presence or absence of distal atherosclerosis (microatheroma; Figure 1C), lipohyalinosis, fibrinoid necrosis or microaneurysms [11, 51, 52, 120, 121, 122, 123, 124, 125 ]. Histopathological measures of NA‐SVD usually rely on semiquantiative scoring of arteriolosclerosis, for example using the criteria in the Vascular Cognitive Impairment Neuropathology Guidelines (VCING). Another metric, the sclerotic index, also been used as a measure of NA‐SVD pathology in brain tissue [126, 127, 128, 129].
We have previously scored the severity of NA‐SVD pathology in tissue sections using a semiquantitative scale that was incorporated into VCING and based on the extent of thickening of the arteriolar walls and narrowing of the lumens [119, 130, 131]. We showed that individuals with severe NA‐SVD had reduced levels of vWF in the white matter, indicative of vessel loss [15, 130]. Perivascular drainage is likely to be impaired (as suggested by the enlarged perivascular spaces [52, 132, 133]). Like CAA, NA‐SVD is associated with lacunar infarcts, haemorrhage and microbleeds but their distribution varies between these two forms of SVD [33, 52, 134, 135] (Figure 1D). Microbleeds tend to be lobar in CAA, and non‐lobar (basal ganglia, internal capsule, thalami) in NA‐SVD [52]. Microbleeds are demonstrable in up to one‐third of AD patients [15, 136, 137, 138 ], and mostly lobar in distribution; however a substantial number are also non‐lobar and associated with WMHs, reflecting the high prevalence of both major types of SVD in AD [13, 14, 15]. It seems likely NA‐SVD reduces cerebrovascular reactivity but supporting data is scarce [33, 139]. Independently of cerebral infarction and CAA, arteriolosclerosis has been linked with limbic predominant age‐related TDP‐43 pathology [11, 46, 140, 141, 142, 143, 144 ], a proteinopathy that often coexists with AD [144, 145].
3. VASCULAR DENSITY AND REMODELLING IN AD
3.1. Changes in vascular density
Most studies on human brain tissue have reported reduced or no significant changes in vascular density (Table 1). Reported reductions in vascular density have tended to be region‐specific and related to disease progression and the presence of disease pathology (Table 1). A reduction in retinal vascular density has also been reported in AD (see section 7 and Table 2). In transgenic mouse and rat models of Aβ accumulation, vascular density is generally decreased (Table 2), though two recent studies reported a transient increase in vascularity, mostly hippocampal, in early disease but a subsequent decline in vascular density with disease progression [146, 147]. This may indicate an early stage angiogenic response that is ultimately ineffective as disease progresses. A few human post‐mortem studies found increased vascular density in AD within the hippocampus [148, 149, 150] (Table 1) and the frontal cortex [151], though several other studies found either no significant change or a reduction in vascularity in these regions (Table 1).
TABLE 1.
Vascular density reported in human patients with AD
Author/year | Change in VD | Region | Method; metric | Stage of disease progression |
---|---|---|---|---|
Fernandez‐Klett et al. (2020) [151] | Increase | Frontal cortex | IHC (CD31); vessel length density | AD |
Burke et al. (2014) [150] | Increase | Hippocampus | IHC (Glut1, ColIV, αSMA); vessel length density | AD |
Desai et al. (2009) [149] | Increase in hippocampus | Multiple regions | Bright field microscopy; branch counts | AD |
Meier‐Ruge et al. (1985) [154] | Increase in capillary volume and length | Multiple regions | Alkaline phosphatase lead‐staining; multiple metrics | AD |
Bell et al. (1981) [148] | Increase | Hippocampus | Alkaline phosphatase lead‐staining; multiple metrics | AD |
Bell et al. (1986) [354] | NS change | Hippocampus and calcarine (visual) cortex | Alkaline phosphatase lead‐staining; vessel length density | AD |
Kawai et al. (1990) [355] | NS change | Hippocampus | IHC (GLUT1 & ColIV labelling); vessel counts | AD |
Bell et al. (1990) [356] | NS change | Visual cortex | Alkaline phosphatase lead‐staining; vessel length density | AD |
Hunter et al. (2012) [155] | NS change | Multiple regions | IHC (ColIV); multiple metrics | AD |
Harris et al. (2018) [357] | NS change | Parietal cortex | ELISA (vWF); vWF mU/mL | AD |
Miners et al. (2017) [277] | NS change | Precuneus | Dot blot (vWF); dot intensity | AD |
Thomas et al. (2015) [358] | NS change | Multiple regions | Dot blot (vWF); dot intensity | AD |
Kirabali et al. (2020) [156] | NS change | Cortex and hippocampus | IF (fluorescent lectin); multiple metrics | AD |
Ding et al. (2020) [276] | NS change | Frontal lobe white matter | IHC and IF (ColIV); vessel length density | AD |
Ding et al. (2021) (285) | NS change | Frontal cortex | IHC and IF (ColIV); vessel length density | AD |
Damodarasamy et al. (2020) [206] | NS change | Frontal and parietal cortices | IHC (CD31 and vWF); vessel area density | AD |
Fischer et al. (1990) [153] | Decrease | Multiple regions | Alkaline phosphatase lead‐staining; branch counts | AD |
Brown et al. (2007) [359] | Decrease | White matter | Alkaline phosphatase lead‐staining; vessel area density | AD |
Lepelletier et al. (2017) [186] | Decrease | Multiple regions | IHC (vWF); vessel area density | Preclinical AD & AD |
Buee et al. (1994) [360] | Decrease | Cortex | IHC (HSPG); vessel area density | AD |
Baloyannis et al. (2012) [161] | Decrease | Multiple regions | Golgi silver staining and electron microscopy; branch counts | AD |
Kitaguchi et al. (2007) [157] | Decrease | Cortex | Gallyas silver staining; branch counts | AD |
van de Kreeke et al. (2019) [332] | Increase | Retina | OCTA | Preclinical AD (Aβ+) |
den Haan et al. (2019) [329] | NS or no change | Retina | OCTA | AD |
Zhang et al. (2019) [319] | Decrease | Retina | OCTA; vessel length density | Early AD and aMCI |
Yoon et al. (2019) [320] | Decrease with AD, not with MCI | Retina | OCTA | MCI & AD |
Jiang et al. (2018) [321] | Decrease | Retina | OCTA; box counting | MCI & AD |
Grewal et al. (2018) [322] | Decrease | Retina | OCTA | AD |
Bulut et al. (2018) [323] | Decrease | Retina | OCTA | AD |
Yan et al. (2021) [324] | Decrease | Retina | OCTA | AD |
Wang et al. (2021) [325] | Decrease | Retina | OCTA | MCI & AD |
Chua et al. (2020) [326] | Decrease | Retina | OCTA | MCI & AD |
Wu et al. (2020) [327] | Decrease | Retina | OCTA | MCI & AD |
Shin et al. (2021) [331] | Decreased; worse in APOE ε4 carriers | Retina | OCTA | MCI |
Abbreviations: AD, Alzheimer's disease; IF, immunofluorescence; IHC, immunohistochemistry; MCI, Mild cognitive impairment; mo, months; NS, no significant; OCTA, optical coherence tomography angiopathy; VD, vascular density.
TABLE 2.
Vascular density reported in AD‐like animal models
Author/year | Change in VD | Animal model | Region | Method; metric | Age |
---|---|---|---|---|---|
Bennett et al. (2018) [42] | Increase | Mouse (P301L) | Cortex | In vivo two‐photon imaging of fluorescein‐conjugated dextran perfused brain; vessel volume density, vessel length density | 2, 9, 12, 15 and 18 mo |
Delafontaine‐Martel et al. (2018) [349] | NS or no change | Mouse (APP/PS1) | Whole brain | Two‐photon imaging of FITC‐gelatin perfused brain; vessel volume density | 2, 4. 5 and 8 mo |
Nikolajsen et al. (2015) [361] | NS or no change | Mouse (APPswe/PS1dE9) | Multiple regions | IHC (CD31); vessel length density | 18 mo |
Paris et al. (2004a) [362] | Decrease | Mouse (Tg4510) | Hippocampus; cyngulate cortex and neocortex | IHC (PECAM‐1); vessel counts | 9 mo |
Herring et al. (2016) [363] | Decrease | Mouse (TgCRND8) | Neocortex and basal ganglia | IHC (laminin); branch counts | 360 d |
Zhang et al. (2019) [364] | Decrease | Mouse (3xTg‐AD) | Neocortex and underlying collateral zone | IF (GLUT‐1); semi‐quantitative scoring | 8 and 18 mo |
Miao et al. (2005) [365] | Decrease | Mouse (Tg‐SwDI) | Cortex, hippocampus, and thalamus | IHC (Collagen IV); vessel counts | 3–24 mo |
Keyvani et al. (2018) [366] | Decrease | Mouse (TgCRND8) | Cortex, hippocampus, and thalamus | IHC (Laminin); vessel area density | 90, 180 & 360 d |
Lee et al. (2005) [367] | Decrease | Mouse (dtg‐APP/PS1) | Corpus callosum | IHC (β‐NADPH); multiple metrics | 7 mo |
Li et al. (2019) [162] | Decrease | Mouse (PS1 / tauP301L / APPSwe) | Whole brain | HFμDI; vessel volume density | 4 and 11 mo |
Zhang et al. (2019) [165] | Decrease | Mouse (APP/PS1) | Whole brain | IF (Nissl); multiple metrics | 2–24 mo |
Kouznetsova et al. (2006) [368] | Decrease | Mouse (Tg2576) | Cortex | IF (Glut1); semi‐quantitative scoring and % capillary load | 4–18 mo |
Giuliani et al. (2019) [146] | Decrease a | Mouse (Tg2576) | Cortex | IF (laminin); vessel volume density | 1–27 mo |
Xu et al. (2020) [147] | Decrease a | Mouse (APP23) | Cortex and hippocampus | MRI; multiple metrics | 3–20 mo |
Stefanova et al. (2018) [369] | Decrease | Rat (OXYS) | Hippocampus | Bright field microscopy; vessel counts | 20 d, 5 mo, 18 mo |
Abbreviations: d, days; H&E, haematoxylin and eosin; HFμDI, high‐frequency micro‐Doppler imaging; IF, immunofluorescence; IHC, immunohistochemistry; mo, months; tg, transgenic; VD, vascular density.
Transient increase in VD observed in young mice.
Advanced age is associated with a decline in vascular density (reviewed here Ref. [152]), making it important to use age‐match controls when assessing vascular changes in the context of AD. The reduced vascular density in AD reported by Fischer and colleagues (1990) may have been confounded by the younger age of the controls (mean 60.8 years) compared to AD cases (mean 84.8 years) [153]. Advanced age is also associated with cerebral atrophy, but in most brain regions this is more marked in AD. Brain atrophy may reduce separation of blood vessels, increasing vascular density without the generation of new vessels. Meier‐Ruge and colleagues (1985) attributed an increase in vascular density in the AD brains they studied to tissue atrophy [154]. However, Hunter and colleagues (2012) reported no change in vascular density in AD, despite tissue atrophy [155], and Kirabali and colleagues (2020) found no difference in vascular density in the frontal cortex and hippocampus in AD despite a reduction in the nearest distance between capillaries, perhaps reflecting parenchymal atrophy [156]. Kitaguchi et al. (2007) observed lower vascular density in AD despite greater atrophy than in controls [157].
Discrepant data on vascular density in AD probably reflect differing methods, metrics, brain regions, disease stage, and confounders such as comorbidity or age. The heterogeneous distribution of tissue atrophy across the brain and between individuals [158, 159, 160] further complicates comparison of vascular density measurements in AD.
3.2. Morphological changes in the vasculature
Numerous morphological abnormalities have been reported in AD and animals modelling aspects of the disease. These include increased vascular tortuosity and looping and kinking in AD [153, 161] and transgenic mice overexpressing APP [162, 163]; irregularities in capillary diameter in AD [157, 161, 164] and APP/PS1 mice [165]; abnormal patterns of branching, fusion and budding of vessels in AD [161] and APP23 mice [157, 163]; and an increase in degenerated ‘string’ vessels in AD [155, 166]. Raspberry‐like clusters of cerebrocortical microvessels that probably reflect an angiogenic response to brain ischaemia were found to be most numerous in VaD but also more numerous in frontotemporal lobar degeneration and AD than in controls [167].
Accumulation of tau may also contribute to vascular abnormalities. Overexpression of tau in mice caused a range of vessel abnormalities including reductions in diameter, the formation of vascular spirals, and altered vascular density [42].
3.3. Non‐productive angiogenesis in AD
The causes of most of the above changes to vascular morphology in AD remain unclear. Brain atrophy is likely to have deformed the morphology of some vessels [168]; however looping, budding, fusion and tapering of vessels may be secondary to angiogenic stimulation [163, 169]. String vessels in AD have been suggested to reflect a cycle of pathological angiogenesis and subsequent endothelial retraction [170] (Figure 1F). Recently reported ‘raspberries’ likely form through angiogenesis in response to tissue hypoxia [167].
Despite evidence of pro‐angiogenic signalling in AD, there is scant evidence of increased vascularization, possibly owing to anti‐angiogenic properties of Aβ [5, 171, 172]. In AD and animal models of Aβ accumulation, non‐productive angiogenesis was shown near amyloid plaques, with an abnormal accumulation of IB4‐positive tip cells and reduced NOTCH signalling. The angiogenic vessels were disassembled by microglial phagocytosis. Non‐productive angiogenesis also occurred in the absence of plaques in mice with inhibited γ‐secretase activity [173].
4. ALTERATIONS IN THE CEREBRAL VASCULAR BASEMENT MEMBRANE IN AD
The ECM provides structural stabilisation to the neurovascular unit by binding cell‐adhesion molecules. It also supports cell migration and differentiation, and facilitates cell signalling [174]. The cerebral vascular basement membrane (CVBM) is a specialised ECM composed of laminin, collagen IV, nidogen, heparan sulphate and proteoglycans. It encloses endothelial cells and pericytes and supports interactions between them, and with astrocytes through their endfeet [175]. Changes to the CVBM in AD have adverse consequences for vascular function and Aβ efflux (reviewed here Refs. [31, 174, 175, 176, 177, 178]). The major alterations to the CVBM in AD include thickening, changes in composition, and Aβ deposition [175].
4.1. CVBM thickening
The CVBM thickens with age, but this process is exacerbated in AD and in animal models of Aβ accumulation [168, 179, 180, 181, 182, 183, 184, 185, 186], tending to be most severe in regions of the brain with higher levels of AD pathology [180, 186]. It is attributable mostly to increased collagen IV [94, 182, 183, 187, 188, 189], although perlecan and fibronectin are also increased [186, 190]. One study found elevated collagen I and III but reduced collagen IV [191]. A few reports describe CVBM thinning in AD microvessels, though those studies focussed on agrin and may reflect decreases in that particular component [192, 193, 194]. Thickening increases the stiffness of the BM, and probably attenuates vascular compliance and neurovascular coupling. Reduced contact between endothelial cells and pericytes [195] may also affect BBB function and the stability of capillaries as a result of CVBM thickening.
4.2. CVBM compositional changes
In addition to the deposition of collagen, increases in perlecan and fibronectin were found in AD microvessels [186] (although not in APOE ε4 mice [196]). These constituents contribute to maintaining the endothelial barrier and mediating cell attachment and function [197, 198]. Plasma fibronectin was increased in AD but reduced in MCI [199, 200]. Laminin α1, β1 and γ1 expression was increased in brain tissue and astrocytes in AD [201, 202] and small laminin peptides were elevated in the CSF [203]. Laminin and other components of the CVBM, including collagen IV and heparan sulphate proteoglycan, also colocalise with amyloid plaques, which reflect CVBM damage [204, 205]. However, both collagen IV and laminin were reportedly reduced in microvessels from AD brain tissue, while laminin was elevated in microvessels with CAA. The same study found that levels of perlecan and fibronectin were unchanged in AD and CAA [206].
Intramural perivascular drainage (IPAD) of interstitial fluid in the brain takes place along the vascular CVBM, particularly those that define the concentric layers of smooth muscle cells and is an important mechanism of Aβ clearance [207, 208]. Pathological remodelling of the CVBM can interfere with this process (Figure 2). Reduced compliance of vessels with thickened CVBMs in normal ageing may explain reduced cardiovascular pulse propagation [209], thought to drive perivascular transport in the brain via its reflection wave [31, 210, 211]. Additionally, elevated levels of perlecan, and fibronectin, owing to normal aging may facilitate the aggregation of soluble Aβ [180]. Additionally, CVBM laminin, which is reduced in ageing and CAA, binds APOE‐Aβ complexes and assists in the efflux of Aβ from the brain. This binding is weaker for APOE ε4‐Aβ than APOE ε3‐Aβ complexes [212]. Astrocytes from ε4‐positive individuals secreted less laminin and collagen IV, and more fibronectin when forming the CVBM [213]. Conversely, ECM laminin and collagen were increased in ε2 carriers [214]. These differences in composition of the CVBM may influence the efficiency of intramural periarterial drainage. Changes in CVBM and ECM composition can also affect Aβ fibrillation and stability. For instance, collagen IV, laminin and nidogen can disrupt the formation of Aβ fibrils [215, 216]. Conversely, proteoglycans such as perlecan and agrin aid Aβ fibril formation and stability [217, 218, 219].
Caution is warranted in interpreting some of the data on immunolabelling components of the CVBM, which is highly cross‐linked. The detection of CVBM components is affected by variability in antigen affinity and accessibility (e.g. in a study on formalin‐fixed tissue, laminin antibodies labelled neurons but not the CVBM unless extensively damaged [220]), in addition to autofluorescence [221].
5. THE NEUROVASCULAR UNIT IN AD
Endothelial cells, mural cells, astrocytes and neurons work in concert in the healthy brain to form the neurovascular unit (NVU), which regulates neurovascular coupling and consequently CBF through the brain (reviewed here Refs. [222, 223]). The CVBM and endothelial tight junctions are important non‐cellular structures of the BBB, which sits within the NVU (Figure 3). NVU dysfunction and BBB barrier disruption in AD are closely associated with one another [223]. Below, we describe various pathologies and dysfunction with a focus on the vascular components of the NVU in AD. Glial cells are also important in the normal function of the NVU and contribute to its disruption in AD. Astrocytes are important for glymphatic drainage of Aβ [224, 225] and microglial neuroinflammation has multiple effects on vascular function, as reviewed elsewhere [226, 227]. However, detailed consideration of the roles of astrocytes and microglia in AD is beyond the scope of the present review.
FIGURE 3.
Structure of the microvascular NVU in health and AD. (A) Structure of NVU in the microvasculature. Endothelial cells form tight junctions that limit the transport of blood constituents. The tight junctions are a major component of the BBB, the integrity of which is further enforced by the basement membrane and pericytes surrounding the endothelium. Astrocytic endfeet cover most of the surface of the microvessel, and form water channels important for glymphatic transport and homeostasis within the brain. Neurons and astrocytes can signal to pericytes to modulate their state of constriction, linking regional CBF to neuronal activity. (B) NVU pathology and dysfunction in AD. Numerous changes in the function and physiology of the NVU have been described in the context of AD (outlined here), many of which are also seen to a lesser extent in normal aging
5.1. Endothelial cells
Reported abnormalities of endothelial cells in AD include mitochondrial damage, increased pinocytic vesicles and lipofuscin [161, 170]; upregulated receptor for advanced glycation end products (RAGE), responsible for transporting Aβ into the brain parenchyma [228, 229, 230]; and downregulated low density lipoprotein receptor‐related protein 1 (LRP‐1) receptor, involved in Aβ clearance into the bloodstream [228, 231] (Figure 2). Lower LRP‐1 levels may reflect diminished expression of MEOX2 [232].
Glucose transport across the endothelium and BBB is impaired in AD. Expression of glucose transporter proteins GLUT‐1 and GLUT‐3 is reduced in the endothelium of microvessels in the cerebral cortex, and hippocampus in AD [233, 234, 235, 236, 237], and GLUT‐1 levels are reduced in circulating brain endothelial cells in mild AD [238]. This may exacerbate Aβ and cerebrovascular pathology. A fall in GLUT‐1 and retraction of astrocytic endfeet preceded widespread Aβ pathology in arcAβ mice [184]. Knockdown of GLUT‐1 in zebrafish caused BBB breakdown and impaired cerebral circulation [239]. In mice over‐expressing APP, GLUT‐1 deficiency led to early cerebral microvascular degeneration, BBB breakdown, reduced CBF, impaired neurovascular coupling, accelerated Aβ accumulation and neurodegeneration [240].
Endothelial cells in AD patients and 5xFAD mice have shortened tight junctions (TJs) [161, 241], important for maintaining BBB integrity. Additionally, there is loss of TJ proteins occludin, claudin‐5 and ZO‐1 in CAA; in vitro experiments suggested that this results from Aβ toxicity to endothelial cells, mediated by the binding of Aβ to endothelial RAGE and induction of oxidative stress [242, 243]. Other studies showed an association of RAGE, Ca2+‐calcineurin signalling and MMP expression, with a decrease in TJ protein levels in endothelial cells [41, 241]. Hypoxia, hypoglycaemia and oxidative stress – conditions prevalent in AD brain tissue – all decrease TJ protein levels in endothelial cells in vitro [244, 245, 246]. Disruption of TJs in AD brains is associated with BBB breakdown and increased immune cell infiltration [247] and in Tg2576 mice, disrupted TJs were linked to angiogenesis [248].
Markers of endothelial activation, including intercellular adhesion molecule‐1 (ICAM‐1) and vascular adhesion molecule‐1 (VCAM‐1), have been reported to be elevated in AD and in animal models of disease [249, 250, 251, 252, 253], either as a result of inflammation or direct exposure to Aβ [253, 254]. This facilitates entry of immune cells into the brain parenchyma where they can exacerbate neuroinflammation and AD pathology [253]. Neutrophil depletion in 3xTg‐AD mice was shown to decrease AD pathology and improve memory [253]. Elevated expression of cell adhesion molecules may also contribute to neutrophil blockage of capillaries observed in APP/PS1 mice [255]. Treatment of these mice with antibody to the neutrophil marker Ly6G increased CBF, possibly by inhibiting the migration of neutrophils towards endothelial inflammation [255].
The glycocalyx on the luminal surface of the endothelium of the brain is an important component of the BBB that shows signs of damage in AD and CAA (reviewed here Ref. [177]). Levels of glycocalyx components hyaluronan and TSG‐6 were increased in microvessels from brain tissue with AD or CAA, which is characteristic of endothelial inflammation and injury [206].
5.2. Pericytes
Pericytes are recruited by endothelial cells in nascent microvessels by signalling between platelet‐derived growth factor‐BB (PDGF‐BB) and its cognate pericyte receptor PDGFRβ. Pericytes regulate an array of processes including BBB functioning, TJ formation, ECM remodelling, angiogenesis, metabolite clearance, and coordinate signalling between other cell types of the NVU [223, 256, 257, 258, 259, 260, 261, 262]. Mice deficient in PDGFRβ‐signalling experience early loss of pericytes, and BBB breakdown [263]. Pericytes can also clear Aβ aggregates via LRP1/ApoE [264] and regulate local CBF through constriction or relaxation of their processes, modulating capillary diameter [265, 266, 267]. Pericyte relaxation is stimulated by neuronal activity, positioning pericytes as important mediators of neurovascular coupling, the process in which CBF is linked to neuronal activity [261, 268, 269]. Embedded within the CVBM, pericytes are able to communicate with endothelial cells through small gaps in the CVBM where the plasma membranes of both cells come into contact [270]. Pericytes can differentiate into various components of the NVU following ischaemic damage [271, 272], suggesting an important role in tissue repair. However, ischaemia was also shown to cause pericyte death ‘in rigor’, irreversibly constricting microvessels and leading to BBB damage [261].
5.2.1. Pericyte dysfunction and loss
Pericyte‐deficient mice manifest an age‐dependent decrease in brain perfusion and neurovascular coupling, associated with BBB breakdown, neurodegeneration and cognitive impairment [260]. These mice also display accelerated Aβ deposition when crossed with animal models of Aβ accumulation [29, 259, 273].
Reduction in pericyte coverage of capillaries was reported in neocortex and hippocampus from AD patients, correlating with BBB breakdown [29, 156, 274]. However, in some studies, capillary pericyte counts were stable in the frontal cortex in AD [151, 275]. This discrepancy could reflect a loss of pericyte processes rather than of pericytes themselves in AD. The number of pericytes was reduced in the frontal white matter in AD, VaD, post‐stroke dementia and mixed dementia [276]. We found a reduction in the pericyte marker PDGFRβ in the precuneus, a region hypoperfused early in AD; the reduction correlated with an increase in fibrinogen indicating BBB breakdown, and with hypoxia and Aβ plaque load [277]. We did not observe a reduction of PDGFRβ in white matter underlying the precuneus. Bourassa et al., measured mural cell markers – PDGFRβ, CD13 and α‐SMA, in microvessels extracted from the parietal cortex of 60 participants in the Religious Orders study and found the marker levels to be reduced in AD in association with TDP‐43 levels [278].
Electron microscopy of the hippocampus, visual, auditory and parietal cortices in AD revealed mitochondrial abnormalities and increased numbers of pinocytic vesicles in pericytes along with a reduction in their overall number; this coincided with a shortening of TJs [161]. In patients with MCI, injury to brain pericytes, assessed by measuring sPDGFRβ in the CSF, was associated with evidence of BBB breakdown on dynamic contrast‐enhanced MRI, and elevated CSF albumin [21, 24, 279]. Pericyte degeneration and BBB breakdown are accelerated in APOE ε4 carriers [280].
Pericytes accumulate Aβ in mouse models of AD, and Aβ1‐40 fibrils reduced pericyte viability and proliferation in vitro [264, 274] (interestingly, Aβ1‐40 monomers had the opposite effect on pericytes, suggesting that the effect of Aβ on pericyte function is aggregation‐dependent, as it is with endothelial cells [274]). Aβ oligomers induced constriction of capillaries by pericytes in human ex vivo tissue and animal models of AD, a process dependent on ROS generation and EDN1 [39]. Capillaries from AD patients were constricted specifically at pericyte locations, with no concomitant change in the diameters of distal arterioles or venules. Pericyte constriction may therefore be chiefly responsible for reduced CBF in AD [39]. BACE‐1 levels increase under hypoxia, owing to a hypoxia‐responsive element in the BACE‐1 promoter, which leads to increased processing of APP into Aβ [281, 282]. This probably results in further pericyte constriction and reduced CBF, forming a vicious cycle [38] (Figure 2). We recently showed that exposure to Aβ interferes with EDN1‐mediated constriction and relaxation of pericytes in vitro [283], which would be expected to impair neurovascular coupling. It is also possible that this interference in the rhythmic constriction and relaxation of mural cells may reduce the effectiveness of clearance of Aβ through IPAD.
6. THE BLOOD‐BRAIN BARRIER IN AD
The structure and function of the BBB have been extensively reviewed [284, 285, 286, 287, 288], as has its disruption in AD [23, 289, 290, 291]. Damage of the BBB was detected preclinically in AD within the hippocampus, independently of the progression of Aβ and tau pathology [24, 289]. BBB breakdown occurs in animal models of Aβ (reviewed here Ref. [292]) and tau accumulation [293], and in AD patients [23, 170, 289]. Blood constituents, including thrombin, fibrinogen, IgG, albumin, and haemoglobin‐derived proteins, can be detected in the brain parenchyma in AD, often in association with amyloid plaques [29, 194, 277, 280, 294, 295, 296, 297]. Albumin levels tend to be elevated in the CSF in MCI and AD [21, 23, 277, 298, 299, 300, 301]. Within the brain parenchyma, plasmin, fibrinogen, thrombin, and albumin can cause neurovascular damage, inflammation, oedema, and ECM degradation [23]. The pathology resulting from plasmin leakage alone was demonstrated by the markedly reduced inflammation and Aβ deposition that followed depletion of plasminogen in Tg6799 mice [302].
An age‐dependent increase in BBB permeability correlates with a rise in the CSF level of a cleaved, soluble form of platelet‐derived growth factor receptor β (sPDGFRβ) shed from damaged pericytes; this is exacerbated in MCI [21]. We found that BBB breakdown (evidenced by accumulation of fibrinogen in post‐mortem tissue from individuals with AD, VaD and mixed dementia) was associated with increased endothelin‐1 (EDN1), more severe hypoperfusion (lower myelin‐associated glycoprotein:proteolipid‐1 ratio), SVD, Aβ and tau [119]. BBB breakdown in AD patients is associated with a reduction in CBF [23, 28, 303, 304, 305]. Tau‐PET imaging revealed a negative correlation between tau pathology and CBF in the temporoparietal regions, exacerbated by the presence of amyloid [43]. APOE ε4 carriers are at increased risk of early BBB breakdown and degeneration of pericytes [193, 194, 280, 297, 306].
Inappropriate activation of MMPs can cause BBB disruption (e.g. following cerebral ischaemia [307]). MMP‐9 knockout mice had reduced infarct volume, neurological deficits and mortality after focal cerebral ischaemia associated with the protection of the BBB [308]. Aβ‐induced activation of MMPs may damage the BBB in AD. In vitro, Aβ1–42 oligomers increased production of RAGE, MMP‐2, MMP‐9, and decreased levels of TJ proteins in bEnd.3 cells [41, 241]. The ensuing BBB disruption could be fully reversed by addition of an anti‐RAGE antibody and partially reversed by a general MMP inhibitor. Similarly, breakdown of the blood‐CSF barrier in mice after exposure to Aβ1–42 oligomers did not occur in the presence of MMP inhibitor or in MMP‐3‐deficient mice [309]. APOE ε4 also drives BBB breakdown, by activating the cyclophilin A‐MMP‐9 pathway in mice and non‐symptomatic human carriers [214, 260, 280, 298]. In vitro, pericytes produced MMP‐9 and migrated in response to TNF‐α [310] (a mediator of neuroinflammation in AD [311]); this could be blocked by anti‐MMP‐9 antibody [310].
Increased activity of MMPs may also contribute to non‐productive angiogenesis in AD (see above). Proteolysis of CVBM components by MMPs is necessary for endothelial migration and sprouting, and tube formation [312, 313, 314].
7. NOVEL INSIGHTS AND METHODS FOR STUDYING THE CEREBRAL VASCULATURE IN AD
7.1. Retinal studies
The study of the retina in AD research was recently reviewed by Shi et al. (2021) [315]. In a mouse model of Aβ accumulation, Shi and colleagues found vascular pathology and pericyte loss alongside retinal Aβ accumulation [316]. They also reported pericyte apoptosis and reduction in PDGFRβ and LRP‐1 associated with Aβ deposition in retinas examined post‐mortem from donors with MCI and AD, mirroring cerebral findings in AD [316]. Additionally, venular abnormalities, microglial activation and astrogliosis were recently demonstrated in the retina coinciding with accumulation of Aβ in an APPNL‐G‐F knock‐in transgenic mouse model [317].
Optical coherence tomography angiography (OCTA) uses laser light reflectance off the surface of haemocytes in motion to produce a map of the microvessels in the retina [318]. Several OCTA studies found a decline in retinal vascular density in AD and MCI (Table 1), associated with morphological anomalies of the vasculature and an increased foveal avascular zone (FAZ) [319, 320, 321, 322, 323, 324, 325, 326, 327, 328]. Two studies reported no difference in retinal vascular density in AD [329, 330] although one did find a thinner choroid in mild AD [330]. Patients with MCI showed a decline in retinal vascular density [319, 321, 325, 326, 327, 331] (with one exception [320]), though to a lesser extent than in AD. Van der Kreeke et al. (2020) reported an increase in retinal vascular density in preclinical AD diagnosed by amyloid‐PET [332]. Changes in retinal vascular density, tortuosity, FAZ area and inner retinal layer thickness are being reported with increasing consistency in AD and other dementias [333, 334]. However, further studies, particularly in preclinical AD and MCI, and with correlative neuropathology, are needed to determine the diagnostic and prognostic value of OCTA in AD [335, 336].
7.2. Single‐cell transcriptomic studies of the cerebral vasculature
Single cell or nuclear RNA‐Seq (snRNA‐Seq) is a powerful tool for transcriptomic analysis of human tissue. Gene studies and snRNA‐Seq profiling have implicated microglia as having a central role in AD pathogenesis (reviewed here Refs. [337, 338, 339, 340]). Of recent snRNA‐Seq studies in AD [171, 341, 342, 343], few have focused on the cerebral vasculature, although recent findings include increased transcription of cytokines [343], angiogenic markers and proteins involved in endothelial antigen presentation in AD [171]. Two recent RNA‐Seq studies were performed specifically on cerebral vascular cells. Song et al. 2020 used laser capture microdissection to isolate microvessels from tissue sections prior to RNA extraction and sequencing [344]. As this study was not snRNA‐Seq, it relied on subsequent analysis to delineate cell‐type specific changes [344]. Yang and colleagues used density centrifugation and strainer capture to enrich microvessels from brain tissue, followed by mashing to release cell nuclei for RNA extraction in a technique they refer to as Vessel Isolation and Nuclei Extraction for Sequencing (VINE‐Seq) [345]. Both of these transcriptomic analyses identified novel putative markers enriched in human pericytes, and documented changes in AD and striking differences between expression of genes in human brain tissue and of their homologs in mice [344, 345]. The VINE‐Seq study found that the expression of many risk genes for AD, identified by GWAS, are highly enriched within vascular cell types in humans whereas in mice most of the homologous genes are expressed by microglia [345]. A post‐GWAS analysis of previous snRNA‐Seq data also found GWAS gene expression to be highly enriched in endothelial cells and pericytes in addition to microglia [346, 347]. These studies highlight the pivotal role of vasculature dysfunction in AD.
7.3. Novel imaging methods in animal models
The numerous recent advances in the imaging of human brains in AD have been well reviewed [23, 348]. Advances in the imaging of the mouse brain vasculature have yielded detailed whole brain atlases of the cerebral vasculature in experimental models of Aβ and tau accumulation. These include the use of iterative sectioning and imaging of the brains of APP/PS1 mice to provide a 3D reconstruction of cerebral vasculature [165, 349], and the demonstration of reduced hippocampal vascular density in a triple‐transgenic mice (PS1M146V, tauP301L, and APPSwe), by ultrasound measurement of cerebral blood flow [162]. Whole brain imaging using tissue clearing methods such as CLARITY has been shown to be effective [350, 351]. Recently, multiphoton imaging and optogenetic manipulation of mural cells in the live mouse brain was demonstrated as a technique for investigating the role of these cells in regulating rCBF and their dysfunction in the context of AD and cerebral ischaemia [352]. Recently, 2D‐optical imaging spectroscopy (2D‐OIS) was used in a J20‐hAPP mouse model of AD to investigate cerebral haemodynamics while measuring neuronal activity with an inserted electrode [353]. In vivo experimental methods such as these are likely to be increasingly important for mechanistic studies of cerebral vascular abnormalities in AD, and for testing novel therapies.
8. CONCLUSION
There is mounting evidence that blood vessels within the brain have altered structure and function from a very early preclinical stage of AD. Structural changes to the larger microvessels (particularly arterioles) include CAA and arteriolosclerosis, but there are also numerous, less immediately obvious, changes to endothelial cells, pericytes and the basement membrane of the microvasculature that reflect the influence of genetic factors, inflammatory mediators, vasoactive peptides, and both direct and indirect endothelial and pericyte responses to Aβ and tau. These structural and physiological alterations to the cerebral vasculature affect vessel maintenance and regeneration, vessel calibre and responsiveness to neuronal metabolic demand, integrity of the BBB, and the metabolism, transport and clearance of many molecules including Aβ. The consequences are a worsening of the reduction and mismatch of brain perfusion in AD, breakdown of the BBB, accumulation of Aβ, parenchymal brain damage, and further damage to the vasculature itself. Transcriptomics of cerebrovascular cells and improved imaging methods of the brain vasculature further highlight the pivotal role of vasculature dysfunction in the development and progression of AD, and the use of the retina as a ‘window into the brain’ may change the way we monitor development and progression of the disease.
CONFLICT OF INTEREST
We have no conflicts of interest to declare.
AUTHOR CONTRIBUTIONS
J. Scott Miners devised the concept and scope of the review. Robert A. Fisher wrote the original draft and constructed the tables and figures. J. Scott Miners and Seth Love edited manuscript and provided guidance on content.
Fisher RA, Miners JS, Love S. Pathological changes within the cerebral vasculature in Alzheimer’s disease: New perspectives. Brain Pathol. 2022;32:e13061. 10.1111/bpa.13061
Funding information
Some of the studies described in this review were supported by funding from Alzheimer's Research UK (ARUK‐SRF‐2019A‐001), Alzheimer's Society (UK)
Contributor Information
Robert A. Fisher, Email: Robbie.fisher@bristol.ac.uk.
Seth Love, Email: Seth.Love@bristol.ac.uk.
DATA AVAILABILITY STATEMENT
Not applicable.
REFERENCES
- 1. Alzheimer’s disease facts and figures. Alzheimers Dement. 2020. Mar;16(3):391–460. [Google Scholar]
- 2. Jellinger KA, Attems J. Prevalence of dementia disorders in the oldest‐old: an autopsy study. Acta Neuropathol. 2010. Apr;119(4):421–33. [DOI] [PubMed] [Google Scholar]
- 3. Brenowitz WD, Hubbard RA, Keene CD, Hawes SE, Longstreth WT, Woltjer RL, et al. Mixed neuropathologies and estimated rates of clinical progression in a large autopsy sample. Alzheimers Dement. 2017. Jun;13(6):654–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Kapasi A, DeCarli C, Schneider JA. Impact of multiple pathologies on the threshold for clinically overt dementia. Acta Neuropathol. 2017. Aug;134(2):171–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Love S, Miners JS. Cerebrovascular disease in ageing and Alzheimer’s disease. Acta Neuropathol. 2016. May;131(5):645–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Kim HW, Hong J, Jeon JC. Cerebral small vessel disease and Alzheimer’s disease: a review. Front Neurol. 2020. Aug 25;11:927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Chalmers K, Wilcock GK, Love S. APOE epsilon 4 influences the pathological phenotype of Alzheimer’s disease by favouring cerebrovascular over parenchymal accumulation of a beta protein. Neuropathol Appl Neurobiol. 2003. Jun;29(3):231–8. [DOI] [PubMed] [Google Scholar]
- 8. Premkumar DR, Cohen DL, Hedera P, Friedland RP, Kalaria RN. Apolipoprotein E‐epsilon4 alleles in cerebral amyloid angiopathy and cerebrovascular pathology associated with Alzheimer’s disease. Am J Pathol. 1996. Jun;148(6):2083–95. [PMC free article] [PubMed] [Google Scholar]
- 9. Love S, Miners S, Palmer J, Chalmers K, Kehoe P. Insights into the pathogenesis and pathogenicity of cerebral amyloid angiopathy. Front Biosci. 2009. Jan 1;14:4778–92. [DOI] [PubMed] [Google Scholar]
- 10. Kalaria RN. The pathology and pathophysiology of vascular dementia. Neuropharmacology. 2018. May;134:226–39. [DOI] [PubMed] [Google Scholar]
- 11. Blevins BL, Vinters HV, Love S, Wilcock DM, Grinberg LT, Schneider JA, et al. Brain arteriolosclerosis. Acta Neuropathol. 2021. Jan;141(1):1–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Kalaria RN, Akinyemi R, Ihara M. Stroke injury, cognitive impairment and vascular dementia. Biochim Biophys Acta. 2016. May;1862(5):915–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Benedictus MR, Goos JDC, Binnewijzend MAA, Muller M, Barkhof F, Scheltens P, et al. Specific risk factors for microbleeds and white matter hyperintensities in Alzheimer’s disease. Neurobiol Aging. 2013. Nov;34(11):2488–94. [DOI] [PubMed] [Google Scholar]
- 14. van der Vlies AE, Goos JDC, Barkhof F, Scheltens P, van der Flier WM. Microbleeds do not affect rate of cognitive decline in Alzheimer disease. Neurology. 2012. Aug 21;79(8):763–9. [DOI] [PubMed] [Google Scholar]
- 15. Love S, Miners JS. Small vessel disease, neurovascular regulation and cognitive impairment: post‐mortem studies reveal a complex relationship, still poorly understood. Clin Sci. 2017. Jun 30;131(14):1579–89. [DOI] [PubMed] [Google Scholar]
- 16. Arvanitakis Z, Capuano AW, Leurgans SE, Bennett DA, Schneider JA. Relation of cerebral vessel disease to Alzheimer’s disease dementia and cognitive function in elderly people: a cross‐sectional study. Lancet Neurol. 2016. Aug;15(9):934–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Iturria‐Medina Y, Sotero RC, Toussaint PJ, Mateos‐Pérez JM, Evans AC. Early role of vascular dysregulation on late‐onset Alzheimer’s disease based on multifactorial data‐driven analysis. Nat Commun. 2016. Sep;7(1):11934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Iturria‐Medina Y, Carbonell FM, Sotero RC, Chouinard‐Decorte F, Evans AC. Multifactorial causal model of brain (dis)organization and therapeutic intervention: application to Alzheimer’s disease. NeuroImage. 2017. May;152:60–77. [DOI] [PubMed] [Google Scholar]
- 19. Ruitenberg A, den Heijer T, Bakker SLM, van Swieten JC, Koudstaal PJ, Hofman A, et al. Cerebral hypoperfusion and clinical onset of dementia: the Rotterdam study. Ann Neurol. 2005. Jun;57(6):789–94. [DOI] [PubMed] [Google Scholar]
- 20. Wolters FJ, Zonneveld HI, Hofman A, van der Lugt A, Koudstaal PJ, Vernooij MW, et al. Cerebral perfusion and the risk of dementia: a population‐based study. Circulation. 2017. Aug 22;136(8):719–28. [DOI] [PubMed] [Google Scholar]
- 21. Montagne A, Barnes S, Sweeney M, Halliday M, Sagare A, Zhao Z, et al. Blood‐brain barrier breakdown in the aging human hippocampus. Neuron. 2015. Jan;85(2):296–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Wardlaw JM, Makin SJ, Valdés Hernández MC, Armitage PA, Heye AK, Chappell FM, et al. Blood‐brain barrier failure as a core mechanism in cerebral small vessel disease and dementia: evidence from a cohort study. Alzheimers Dement. 2017. Jun;13(6):634–43. [Google Scholar]
- 23. Sweeney MD, Sagare AP, Zlokovic BV. Blood–brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat Rev Neurol. 2018;14(3):133–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Nation DA, Sweeney MD, Montagne A, Sagare AP, D’Orazio LM, Pachicano M, et al. Blood–brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nat Med. 2019. Feb;25(2):270–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Benzinger TLS, Blazey T, Jack CR, Koeppe RA, Su Y, Xiong C, et al. Regional variability of imaging biomarkers in autosomal dominant Alzheimer’s disease. Proc Natl Acad Sci U S A. 2013. Nov 19;110(47):E4502–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. McDade E, Kim A, James J, Sheu LK, Kuan D C‐H, Minhas D, et al. Cerebral perfusion alterations and cerebral amyloid in autosomal dominant Alzheimer disease. Neurology. 2014. Aug 19;83(8):710–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. McDade E, Wang G, Gordon BA, Hassenstab J, Benzinger TLS, Buckles V, et al. Longitudinal cognitive and biomarker changes in dominantly inherited Alzheimer disease. Neurology. 2018. Oct 2;91(14):e1295–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. van de Haar HJ, Jansen JFA, van Osch MJP, van Buchem MA, Muller M, Wong SM, et al. Neurovascular unit impairment in early Alzheimer’s disease measured with magnetic resonance imaging. Neurobiol Aging. 2016. Sep;45:190–6. [DOI] [PubMed] [Google Scholar]
- 29. Sengillo JD, Winkler EA, Walker CT, Sullivan JS, Johnson M, Zlokovic BV. Deficiency in mural vascular cells coincides with blood‐brain barrier disruption in Alzheimer’s disease. Brain Pathol. 2013. May;23(3):303–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Howe MD, McCullough LD, Urayama A. The role of basement membranes in cerebral amyloid angiopathy. Front Physiol. 2020. Nov 25;11:601320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Morris AWJ, Carare RO, Schreiber S, Hawkes CA. The cerebrovascular basement membrane: role in the clearance of β‐amyloid and cerebral amyloid angiopathy. Front Aging Neurosci. 2014. Sep 19;6:251. 10.3389/fnagi.2014.00251/abstract [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Agarwal N, Carare RO. Cerebral vessels: an overview of anatomy, physiology, and role in the drainage of fluids and solutes. Front Neurol. 2021. Jan 13;11:611485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Wardlaw JM, Benveniste H, Nedergaard M, Zlokovic BV, Mestre H, Lee H, et al. Perivascular spaces in the brain: anatomy, physiology and pathology. Nat Rev Neurol. 2020. Mar 15;16(3):137–53. [DOI] [PubMed] [Google Scholar]
- 34. Kanekiyo T, Bu G. The low‐density lipoprotein receptor‐related protein 1 and amyloid‐β clearance in Alzheimer’s disease. Front Aging Neurosci. 2014. May 20;6:93. 10.3389/fnagi.2014.00093/abstract [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Yu Y, Ye RD. Microglial Aβ receptors in Alzheimer’s disease. Cell Mol Neurobiol. 2015. Jan;35(1):71–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Ramanathan A, Nelson AR, Sagare AP, Zlokovic BV. Impaired vascular‐mediated clearance of brain amyloid beta in Alzheimer’s disease: the role, regulation and restoration of LRP1. Front Aging Neurosci. 2015. Jul 15;7:136. 10.3389/fnagi.2015.00136/abstract [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Shinohara M, Tachibana M, Kanekiyo T, Bu G. Role of LRP1 in the pathogenesis of Alzheimer’s disease: evidence from clinical and preclinical studies. J Lipid Res. 2017. Jul;58(7):1267–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Korte N, Nortley R, Attwell D. Cerebral blood flow decrease as an early pathological mechanism in Alzheimer’s disease. Acta Neuropathol. 2020. Dec;140(6):793–810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Nortley R, Korte N, Izquierdo P, Hirunpattarasilp C, Mishra A, Jaunmuktane Z, et al. Amyloid β oligomers constrict human capillaries in Alzheimer’s disease via signaling to pericytes. Science. 2019. Jul;365(6450):eaav9518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Dietrich HH, Xiang C, Han BH, Zipfel GJ, Holtzman DM. Soluble amyloid‐β, effect on cerebral arteriolar regulation and vascular cells. Mol Neurodegener. 2010;5(1):15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Wan W, Cao L, Liu L, Zhang C, Kalionis B, Tai X, et al. Aβ1–42 oligomer‐induced leakage in an in vitro blood–brain barrier model is associated with up‐regulation of RAGE and metalloproteinases, and down‐regulation of tight junction scaffold proteins. J Neurochem. 2015. Jul;134(2):382–93. [DOI] [PubMed] [Google Scholar]
- 42. Bennett RE, Robbins AB, Hu M, Cao X, Betensky RA, Clark T, et al. Tau induces blood vessel abnormalities and angiogenesis‐related gene expression in P301L transgenic mice and human Alzheimer’s disease. Proc Natl Acad Sci U S A. 2018. Feb;115(6):E1289–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Albrecht D, Isenberg AL, Stradford J, Monreal T, Sagare A, Pachicano M, et al. Associations between vascular function and tau PET are associated with global cognition and amyloid. J Neurosci. 2020. Oct 28;40(44):8573–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Michalicova A, Majerova P, Kovac A. Tau protein and its role in blood‐brain barrier dysfunction. Front Mol Neurosci. 2020;13:570045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Wennberg AM, Whitwell JL, Tosakulwong N, Weigand SD, Murray ME, Machulda MM, et al. The influence of tau, amyloid, alpha‐synuclein, TDP‐43, and vascular pathology in clinically normal elderly individuals. Neurobiol Aging. 2019. May;77:26–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Katsumata Y, Fardo DW, Kukull WA, Nelson PT. Dichotomous scoring of TDP‐43 proteinopathy from specific brain regions in 27 academic research centers: associations with Alzheimer’s disease and cerebrovascular disease pathologies. Acta Neuropathol Commun. 2018. Dec 19;6(1):142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. de la Torre JC. Cerebral perfusion enhancing interventions: a new strategy for the prevention of Alzheimer dementia. Brain Pathol. 2016. Sep;26(5):618–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Zhang A‐J, Yu X‐J, Wang M. The clinical manifestations and pathophysiology of cerebral small vessel disease. Neurosci Bull. 2010. Jun;26(3):257–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Gorelick PB, Scuteri A, Black SE, DeCarli C, Greenberg SM, Iadecola C, et al. Vascular contributions to cognitive impairment and dementia: a statement for healthcare professionals from the american heart association/american stroke association. Stroke. 2011. Sep;42(9):2672–713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Debette S, Schilling S, Duperron M‐G, Larsson SC, Markus HS. Clinical significance of magnetic resonance imaging markers of vascular brain injury: a systematic review and meta‐analysis. JAMA Neurol. 2019. Jan 1;76(1):81–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Pantoni L. Cerebral small vessel disease: from pathogenesis and clinical characteristics to therapeutic challenges. Lancet Neurol. 2010. Jul;9(7):689–701. [DOI] [PubMed] [Google Scholar]
- 52. Charidimou A, Pantoni L, Love S. The concept of sporadic cerebral small vessel disease: a road map on key definitions and current concepts. Int J Stroke. 2016. Jan 1;11(1):6–18. [DOI] [PubMed] [Google Scholar]
- 53. Kalaria RN, Sepulveda‐Falla D. Cerebral small vessel disease in sporadic and familial Alzheimer disease. Am J Pathol. 2021. Nov;191(11):1888–905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Schmidt R, Grazer A, Enzinger C, Ropele S, Homayoon N, Pluta‐Fuerst A, et al. MRI‐detected white matter lesions: do they really matter? J Neural Transm Vienna Austria. 2011. May;118(5):673–81. [DOI] [PubMed] [Google Scholar]
- 55. Charidimou A, Farid K, Baron J‐C. Amyloid‐PET in sporadic cerebral amyloid angiopathy: a diagnostic accuracy meta‐analysis. Neurology. 2017. Oct 3;89(14):1490–8. [DOI] [PubMed] [Google Scholar]
- 56. Greenberg SM, Bacskai BJ, Hernandez‐Guillamon M, Pruzin J, Sperling R, van Veluw SJ. Cerebral amyloid angiopathy and Alzheimer disease — one peptide, two pathways. Nat Rev Neurol. 2020. Jan;16(1):30–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Wardlaw JM, Smith C, Dichgans M. Small vessel disease: mechanisms and clinical implications. Lancet Neurol. 2019. Jul;18(7):684–96. [DOI] [PubMed] [Google Scholar]
- 58. Love S, Nicoll JAR, Hughes A, Wilcock GK. APOE and cerebral amyloid angiopathy in the elderly. NeuroReport. 2003. Aug;14(11):1535–6. [DOI] [PubMed] [Google Scholar]
- 59. Keage HAD, Carare RO, Friedland RP, Ince PG, Love S, Nicoll JA, et al. Population studies of sporadic cerebral amyloid angiopathy and dementia: a systematic review. BMC Neurol. 2009. Jan 13;9:3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Esiri MM, Wilcock GK. Cerebral amyloid angiopathy in dementia and old age. J Neurol Neurosurg Psychiatry. 1986. Nov;49(11):1221–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Jellinger KA. Alzheimer disease and cerebrovascular pathology: an update. J Neural Transm. 2002;109(5):813–36. [DOI] [PubMed] [Google Scholar]
- 62. Boyle PA, Yu L, Nag S, Leurgans S, Wilson RS, Bennett DA, et al. Cerebral amyloid angiopathy and cognitive outcomes in community‐based older persons. Neurology. 2015. Dec 1;85(22):1930–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Love S, Chalmers K, Ince P, Esiri M, Attems J, Jellinger K, et al. Development, appraisal, validation and implementation of a consensus protocol for the assessment of cerebral amyloid angiopathy in post‐mortem brain tissue. Am J Neurodegener Dis. 2014;3(1):19–32. [PMC free article] [PubMed] [Google Scholar]
- 64. Wharton SB, Wang D, Parikh C, Matthews FE, Brayne C, Ince PG. Epidemiological pathology of Aβ deposition in the ageing brain in CFAS: addition of multiple Aβ‐derived measures does not improve dementia assessment using logistic regression and machine learning approaches. Acta Neuropathol Commun. 2019. Dec;7(1):198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Biffi A, Sonni A, Anderson CD, Kissela B, Jagiella JM, Schmidt H, et al. Variants at APOE influence risk of deep and lobar intracerebral hemorrhage. Ann Neurol. 2010. Dec;68(6):934–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Olichney JM, Hansen LA, Galasko D, Saitoh T, Hofstetter CR, Katzman R, 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. 1996. Jul;47(1):190–6. [DOI] [PubMed] [Google Scholar]
- 67. Warzok RW, Kessler C, Apel G, Schwarz A, Egensperger R, Schreiber D, et al. Apolipoprotein E4 promotes incipient Alzheimer pathology in the elderly. Alzheimer Dis Assoc Disord. 1998. Mar;12(1):33–9. [DOI] [PubMed] [Google Scholar]
- 68. Thal DR, Ghebremedhin E, Rüb U, Yamaguchi H, Del Tredici K, Braak H. Two types of sporadic cerebral amyloid angiopathy. J Neuropathol Exp Neurol. 2002. Mar;61(3):282–93. [DOI] [PubMed] [Google Scholar]
- 69. Yu L, Boyle PA, Nag S, Leurgans S, Buchman AS, Wilson RS, et al. APOE and cerebral amyloid angiopathy in community‐dwelling older persons. Neurobiol Aging. 2015. Nov;36(11):2946–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Thal DR, Papassotiropoulos A, Saido TC, Griffin WST, Mrak RE, Kölsch H, et al. Capillary cerebral amyloid angiopathy identifies a distinct APOE epsilon4‐associated subtype of sporadic Alzheimer’s disease. Acta Neuropathol. 2010. Aug;120(2):169–83. [DOI] [PubMed] [Google Scholar]
- 71. Mäkelä M, Paetau A, Polvikoski T, Myllykangas L, Tanskanen M. Capillary amyloid‐β protein deposition in a population‐based study (Vantaa 85+). J Alzheimers Dis. 2015. Sep 28;49(1):149–57. [DOI] [PubMed] [Google Scholar]
- 72. Gkanatsiou E, Portelius E, Toomey CE, Blennow K, Zetterberg H, Lashley T, et al. A distinct brain beta amyloid signature in cerebral amyloid angiopathy compared to Alzheimer’s disease. Neurosci Lett. 2019. May;701:125–31. [DOI] [PubMed] [Google Scholar]
- 73. Miller DL, Papayannopoulos IA, Styles J, Bobin SA, Lin YY, Biemann K, et al. Peptide compositions of the cerebrovascular and senile plaque core amyloid deposits of Alzheimer’s disease. Arch Biochem Biophys. 1993. Feb 15;301(1):41–52. [DOI] [PubMed] [Google Scholar]
- 74. Suzuki N, Iwatsubo T, Odaka A, Ishibashi Y, Kitada C, Ihara Y. High tissue content of soluble beta 1–40 is linked to cerebral amyloid angiopathy. Am J Pathol. 1994. Aug;145(2):452–60. [PMC free article] [PubMed] [Google Scholar]
- 75. Gravina SA, Ho L, Eckman CB, Long KE, Otvos L, Younkin LH, 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. 1995. Mar 31;270(13):7013–6. [DOI] [PubMed] [Google Scholar]
- 76. Kakuda N, Miyasaka T, Iwasaki N, Nirasawa T, Wada‐Kakuda S, Takahashi‐Fujigasaki J, et al. Distinct deposition of amyloid‐β species in brains with Alzheimer’s disease pathology visualized with MALDI imaging mass spectrometry. Acta Neuropathol Commun. 2017. Oct 16;5(1):73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Roher AE, Lowenson JD, Clarke S, Woods AS, Cotter RJ, Gowing E, 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. 1993. Nov 15;90(22):10836–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Attems J, Lintner F, Jellinger KA. Amyloid beta peptide 1–42 highly correlates with capillary cerebral amyloid angiopathy and Alzheimer disease pathology. Acta Neuropathol. 2004. Apr;107(4):283–91. [DOI] [PubMed] [Google Scholar]
- 79. Biffi A, Greenberg SM. Cerebral amyloid angiopathy: a systematic review. J Clin Neurol. 2011. Mar;7(1):1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Elder GA, Gama Sosa MA, De Gasperi R, Dickstein DL, Hof PR. Presenilin transgenic mice as models of Alzheimer’s disease. Brain Struct Funct. 2010. Mar;214(2–3):127–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Jäkel L, Van Nostrand WE, Nicoll JAR, Werring DJ, Verbeek MM. Animal models of cerebral amyloid angiopathy. Clin Sci. 2017. Oct 15;131(19):2469–88. [DOI] [PubMed] [Google Scholar]
- 82. Revesz T, Holton JL, Lashley T, Plant G, Rostagno A, Ghiso J, et al. Sporadic and familial cerebral amyloid angiopathies. Brain Pathol. 2002. Jul;12(3):343–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Keable A, Fenna K, Yuen HM, Johnston DA, Smyth NR, Smith C, et al. Deposition of amyloid β in the walls of human leptomeningeal arteries in relation to perivascular drainage pathways in cerebral amyloid angiopathy. Biochim Biophys Acta Mol Basis Dis. 2016. May;1862(5):1037–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Mandybur TI. Cerebral amyloid angiopathy: the vascular pathology and complications. J Neuropathol Exp Neurol. 1986. Jan;45(1):79–90. [PubMed] [Google Scholar]
- 85. Vonsattel JPG, Myers RH, Tessa Hedley‐Whyte E, Ropper AH, Bird ED, Richardson EP. Cerebral amyloid angiopathy without and with cerebral hemorrhages: a comparative histological study. Ann Neurol. 1991. Nov;30(5):637–49. [DOI] [PubMed] [Google Scholar]
- 86. Greenberg SM, Nandigam RNK, Delgado P, Betensky RA, Rosand J, Viswanathan A, et al. Microbleeds versus macrobleeds: evidence for distinct entities. Stroke. 2009. Jul;40(7):2382–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Maeda A, Yamada M, Itoh Y, Otomo E, Hayakawa M, Miyatake T. Computer‐assisted three‐dimensional image analysis of cerebral amyloid angiopathy. Stroke. 1993. Dec;24(12):1857–64. [DOI] [PubMed] [Google Scholar]
- 88. Yamada M. Cerebral amyloid angiopathy: emerging concepts. J Stroke. 2015. Jan;17(1):17–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. McCarron MO, Nicoll JA, Stewart J, Ironside JW, Mann DM, Love S, et al. The apolipoprotein E epsilon2 allele and the pathological features in cerebral amyloid angiopathy‐related hemorrhage. J Neuropathol Exp Neurol. 1999. Jul;58(7):711–8. [DOI] [PubMed] [Google Scholar]
- 90. Wu J‐J, Yao M, Ni J. Cerebral amyloid angiopathy‐related inflammation: current status and future implications. Chin Med J. 2021. Feb 23;134(6):646–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Chwalisz BK. Cerebral amyloid angiopathy and related inflammatory disorders. J Neurol Sci. 2021. May 15;424:117425. [DOI] [PubMed] [Google Scholar]
- 92. Auriel E, Charidimou A, Gurol ME, Ni J, Van Etten ES, Martinez‐Ramirez S, et al. Validation of clinicoradiological criteria for the diagnosis of cerebral amyloid angiopathy‐related inflammation. JAMA Neurol. 2016. Feb;73(2):197–202. [DOI] [PubMed] [Google Scholar]
- 93. van Veluw SJ, Frosch MP, Scherlek AA, Lee D, Greenberg SM, Bacskai BJ. In vivo characterization of spontaneous microhemorrhage formation in mice with cerebral amyloid angiopathy. J Cereb Blood Flow Metab. 2021. Jan;41(1):82–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Magaki S, Tang Z, Tung S, Williams CK, Lo D, Yong WH, et al. The effects of cerebral amyloid angiopathy on integrity of the blood‐brain barrier. Neurobiol Aging. 2018. Oct;70:70–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Richard E, Carrano A, Hoozemans JJ, van Horssen J, van Haastert ES, Eurelings LS, et al. Characteristics of dyshoric capillary cerebral amyloid angiopathy. J Neuropathol Exp Neurol. 2010. Nov;69(11):1158–67. [DOI] [PubMed] [Google Scholar]
- 96. Attems J, Jellinger K, Thal DR, Van Nostrand W. Review: Sporadic cerebral amyloid angiopathy: cerebral amyloid angiopathy. Neuropathol Appl Neurobiol. 2011. Feb;37(1):75–93. [DOI] [PubMed] [Google Scholar]
- 97. Charidimou A, Perosa V, Frosch MP, Scherlek AA, Greenberg SM, van Veluw SJ. Neuropathological correlates of cortical superficial siderosis in cerebral amyloid angiopathy. Brain. 2020. Nov 1;143(11):3343–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Sharma R, Dearaugo S, Infeld B, O’Sullivan R, Gerraty RP. Cerebral amyloid angiopathy: review of clinico‐radiological features and mimics. J Med Imaging Radiat Oncol. 2018. Aug;62(4):451–63. [DOI] [PubMed] [Google Scholar]
- 99. Chen Y, Rabson AB, Gorski DH. MEOX2 regulates nuclear factor‐κB activity in vascular endothelial cells through interactions with p65 and IκBβ. Cardiovasc Res. 2010. Sep;87(4):723–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Peca S, McCreary CR, Donaldson E, Kumarpillai G, Shobha N, Sanchez K, et al. Neurovascular decoupling is associated with severity of cerebral amyloid angiopathy. Neurology. 2013. Nov 5;81(19):1659–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Dumas A, Dierksen GA, Gurol ME, Halpin A, Martinez‐Ramirez S, Schwab K, et al. Functional magnetic resonance imaging detection of vascular reactivity in cerebral amyloid angiopathy. Ann Neurol. 2012. Jul;72(1):76–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Switzer AR, McCreary C, Batool S, Stafford RB, Frayne R, Goodyear BG, et al. Longitudinal decrease in blood oxygenation level dependent response in cerebral amyloid angiopathy. NeuroImage Clin. 2016;11:461–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. van Opstal AM, van Rooden S, van Harten T, Ghariq E, Labadie G, Fotiadis P, et al. Cerebrovascular function in presymptomatic and symptomatic individuals with hereditary cerebral amyloid angiopathy: a case‐control study. Lancet Neurol. 2017. Feb;16(2):115–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Fotiadis P, van Rooden S, van der Grond J, Schultz A, Martinez‐Ramirez S, Auriel E, et al. Cortical atrophy in patients with cerebral amyloid angiopathy: a case‐control study. Lancet Neurol. 2016. Jul;15(8):811–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Drenth N, van der Grond J, Rombouts SARB, van Buchem MA, Terwindt GM, Wermer MJH, et al. Cerebral amyloid angiopathy is associated with decreased functional brain connectivity. NeuroImage Clin. 2021;29:102546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Weller RO, Preston SD, Subash M, Carare RO. Cerebral amyloid angiopathy in the aetiology and immunotherapy of Alzheimer disease. Alzheimers Res Ther. 2009. Oct 12;1(2):6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Herzig MC, Nostrand WE, Jucker M. Mechanism of cerebral β‐amyloid angiopathy: murine and cellular models. Brain Pathol. 2006. Jan;16(1):40–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Roher AE, Kuo Y‐M, Esh C, Knebel C, Weiss N, Kalback W, et al. Cortical and leptomeningeal cerebrovascular amyloid and white matter pathology in Alzheimer’s disease. Mol Med. 2003. Mar;9(3–4):112–22. [PMC free article] [PubMed] [Google Scholar]
- 109. 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. 2008;18(2):253–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Weller RO, Hawkes CA, Kalaria RN, Werring DJ, Carare RO. White matter changes in dementia: role of impaired drainage of interstitial fluid. Brain Pathol. 2015. Jan;25(1):63–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Vogelgesang S, Warzok RW, Cascorbi I, Kunert‐Keil C, Schroeder E, Kroemer HK, et al. The role of P‐glycoprotein in cerebral amyloid angiopathy; implications for the early pathogenesis of Alzheimer’s disease. Curr Alzheimer Res. 2004. May;1(2):121–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Pahnke J, Wolkenhauer O, Krohn M, Walker LC. Clinico‐pathologic function of cerebral ABC transporters – implications for the pathogenesis of Alzheimer’s disease. Curr Alzheimer Res. 2008. Aug;5(4):396–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Miners JS, Van Helmond Z, Chalmers K, Wilcock G, Love S, Kehoe PG. Decreased expression and activity of neprilysin in Alzheimer disease are associated with cerebral amyloid angiopathy. J Neuropathol Exp Neurol. 2006. Oct;65(10):1012–21. [DOI] [PubMed] [Google Scholar]
- 114. Miners JS, Kehoe P, Love S. Neprilysin protects against cerebral amyloid angiopathy and Aβ‐induced degeneration of cerebrovascular smooth muscle cells. Brain Pathol. 2011. Sep;21(5):594–605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. 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. 2019;11:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Meyer JS, Xu G, Thornby J, Chowdhury MH, Quach M. Is mild cognitive impairment prodromal for vascular dementia like Alzheimer’s disease? Stroke. 2002. Aug;33(8):1981–5. [DOI] [PubMed] [Google Scholar]
- 117. Meyer JS, Huang J, Chowdhury M. MRI abnormalities associated with mild cognitive impairments of vascular (VMCI) versus neurodegenerative (NMCI) types prodromal for vascular and Alzheimer’s dementias. Curr Alzheimer Res. 2005. Dec;2(5):579–85. [DOI] [PubMed] [Google Scholar]
- 118. Marini S, Anderson CD, Rosand J. Genetics of cerebral small vessel disease. Stroke. 2020. Jan;51(1):12–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Tayler H, Miners JS, Güzel Ö, MacLachlan R, Love S. Mediators of cerebral hypoperfusion and blood‐brain barrier leakiness in Alzheimer’s disease, vascular dementia and mixed dementia. Brain Pathol. 2021. Mar 29;31(4):e12935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Lammie GA. Hypertensive cerebral small vessel disease and stroke. Brain Pathol. 2002. Jul;12(3):358–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Cole FM, Yates P. Intracerebral microaneurysms and small cerebrovascular lesions. Brain J Neurol. 1967. Dec;90(4):759–68. [DOI] [PubMed] [Google Scholar]
- 122. Feigin I, Prose P. Hypertensive fibrinoid arteritis of the brain and gross cerebral hemorrhage: a form of “hyalinosis”. Arch Neurol. 1959. Jul;1:98–110. [DOI] [PubMed] [Google Scholar]
- 123. Rosenblum WI. Fibrinoid necrosis of small brain arteries and arterioles and miliary aneurysms as causes of hypertensive hemorrhage: a critical reappraisal. Acta Neuropathol. 2008. Oct;116(4):361–9. [DOI] [PubMed] [Google Scholar]
- 124. Fisher CM. Pathological observations in hypertensive cerebral hemorrhage. J Neuropathol Exp Neurol. 1971. Jul;30(3):536–50. [DOI] [PubMed] [Google Scholar]
- 125. Takebayashi S, Kaneko M. Electron microscopic studies of ruptured arteries in hypertensive intracerebral hemorrhage. Stroke. 1983. Jan;14(1):28–36. [DOI] [PubMed] [Google Scholar]
- 126. Hase Y, Polvikoski TM, Firbank MJ, Craggs LJL, Hawthorne E, Platten C, et al. Small vessel disease pathological changes in neurodegenerative and vascular dementias concomitant with autonomic dysfunction. Brain Pathol. 2020. Jan;30(1):191–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Giwa MO, Williams J, Elderfield K, Jiwa NS, Bridges LR, Kalaria RN, et al. Neuropathologic evidence of endothelial changes in cerebral small vessel disease. Neurology. 2012. Jan 17;78(3):167–74. [DOI] [PubMed] [Google Scholar]
- 128. McAleese KE, Firbank M, Dey M, Colloby SJ, Walker L, Johnson M, et al. Cortical tau load is associated with white matter hyperintensities. Acta Neuropathol Commun. 2015. Sep;30(3):60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Craggs LJL, Hagel C, Kuhlenbaeumer G, Borjesson‐Hanson A, Andersen O, Viitanen M, et al. Quantitative vascular pathology and phenotyping familial and sporadic cerebral small vessel diseases. Brain Pathol. 2013. Sep;23(5):547–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Barker R, Ashby EL, Wellington D, Barrow VM, Palmer JC, Kehoe PG, et al. Pathophysiology of white matter perfusion in Alzheimer’s disease and vascular dementia. Brain. 2014. Mar;137(5):1524–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Miners JS, Palmer JC, Love S. Pathophysiology of hypoperfusion of the precuneus in early Alzheimer’s disease. Brain Pathol. 2015. Oct;26(4):533–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Braffman BH, Zimmerman RA, Trojanowski JQ, Gonatas NK, Hickey WF, Schlaepfer WW. Brain MR: pathologic correlation with gross and histopathology. 1. Lacunar infarction and Virchow‐Robin spaces. AJR Am J Roentgenol. 1988. Sep;151(3):551–8. [DOI] [PubMed] [Google Scholar]
- 133. van Swieten JC, van den Hout JH, van Ketel BA, Hijdra A, Wokke JH, van Gijn J. Periventricular lesions in the white matter on magnetic resonance imaging in the elderly. A morphometric correlation with arteriolosclerosis and dilated perivascular spaces. Brain. J Neurol. 1991. Apr;114(Pt 2):761–74. [DOI] [PubMed] [Google Scholar]
- 134. Fisher CM. The arterial lesions underlying lacunes. Acta Neuropathol. 1968. Dec 18;12(1):1–15. [DOI] [PubMed] [Google Scholar]
- 135. Fisher CM. Hypertensive cerebral hemorrhage. demonstration of the source of bleeding. J Neuropathol Exp Neurol. 2003. Jan 1;62(1):104–7. [DOI] [PubMed] [Google Scholar]
- 136. Sepehry AA, Lang D, Hsiung G‐Y, Rauscher A. Prevalence of brain microbleeds in alzheimer disease: a systematic review and meta‐analysis on the influence of neuroimaging techniques. Am J Neuroradiol. 2016. Feb;37(2):215–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Pettersen JA, Sathiyamoorthy G, Gao F‐Q, Szilagyi G, Nadkarni NK, St George‐Hyslop P, et al. Microbleed topography, leukoaraiosis, and cognition in probable Alzheimer disease from the Sunnybrook dementia study. Arch Neurol. 2008. Jun;65(6):790–5. [DOI] [PubMed] [Google Scholar]
- 138. Cordonnier C, van der Flier WM. Brain microbleeds and Alzheimer’s disease: innocent observation or key player? Brain J Neurol. 2011. Feb;134(Pt 2):335–44. [DOI] [PubMed] [Google Scholar]
- 139. Blair GW, Doubal FN, Thrippleton MJ, Marshall I, Wardlaw JM. Magnetic resonance imaging for assessment of cerebrovascular reactivity in cerebral small vessel disease: a systematic review. J Cereb Blood Flow Metab. 2016. May 1;36(5):833–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Besser LM, Teylan MA, Nelson PT. Limbic predominant age‐related TDP‐43 encephalopathy (LATE): clinical and neuropathological associations. J Neuropathol Exp Neurol. 2020. Mar 1;79(3):305–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Besser LM, Kukull WA, Teylan MA, Bigio EH, Cairns NJ, Kofler JK, et al. The Revised National Alzheimer’s Coordinating Center’s Neuropathology Form—available data and new analyses. J Neuropathol Exp Neurol. 2018. Aug 1;77(8):717–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Neltner JH, Abner EL, Jicha GA, Schmitt FA, Patel E, Poon LW, et al. Brain pathologies in extreme old age. Neurobiol Aging. 2016. Jan 1;37:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Neltner JH, Abner EL, Baker S, Schmitt FA, Kryscio RJ, Jicha GA, et al. Arteriolosclerosis that affects multiple brain regions is linked to hippocampal sclerosis of ageing. Brain. 2014. Jan 1;137(1):255–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Nelson PT, Dickson DW, Trojanowski JQ, Jack CR, Boyle PA, Arfanakis K, et al. Limbic‐predominant age‐related TDP‐43 encephalopathy (LATE): consensus working group report. Brain. 2019. Jun 1;142(6):1503–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. James BD, Wilson RS, Boyle PA, Trojanowski JQ, Bennett DA, Schneider JA. TDP‐43 stage, mixed pathologies, and clinical Alzheimer’s‐type dementia. Brain. 2016. Nov 1;139(11):2983–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Giuliani A, Sivilia S, Baldassarro VA, Gusciglio M, Lorenzini L, Sannia M, et al. Age‐related changes of the neurovascular unit in the cerebral cortex of Alzheimer disease mouse models: a neuroanatomical and molecular study. J Neuropathol Exp Neurol. 2019. Feb;78(2):101–12. [DOI] [PubMed] [Google Scholar]
- 147. Xu X, Meng T, Wen Q, Tao M, Wang P, Zhong K, et al. Dynamic changes in vascular size and density in transgenic mice with Alzheimer’s disease. Aging. 2020. Sep;12(17):17224–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Bell MA, Ball MJ. Morphometric comparison of hippocampal microvasculature in ageing and demented people: diameters and densities. Acta Neuropathol. 1981. Dec;53(4):299–318. [DOI] [PubMed] [Google Scholar]
- 149. Desai BS, Schneider JA, Li J‐L, Carvey PM, Hendey B. Evidence of angiogenic vessels in Alzheimer’s disease. J Neural Transm. 2009;116(5):587–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Burke MJC, Nelson L, Slade JY, Oakley AE, Khundakar AA, Kalaria RN. Morphometry of the hippocampal microvasculature in post‐stroke and age‐related dementias. Neuropathol Appl Neurobiol. 2014. Apr;40(3):284–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Fernandez‐Klett F, Brandt L, Fernández‐Zapata C, Abuelnor B, Middeldorp J, Sluijs JA, et al. Denser brain capillary network with preserved pericytes in Alzheimer’s disease. Brain Pathol. 2020. Nov;30(6):1071–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Ambrose CT. A therapeutic approach for senile dementias: neuroangiogenesis. J Alzheimers Dis. 2015;43:1–17. [DOI] [PubMed] [Google Scholar]
- 153. Fischer VW, Siddiqi A, Yusufaly Y. Altered angioarchitecture in selected areas of brains with Alzheimer’s disease. Acta Neuropathol. 1990;79(6):672–9. [DOI] [PubMed] [Google Scholar]
- 154. Meier‐Ruge W, Ulrich J, Stähelin HB. Morphometric investigation of nerve cells, neuropil and senile plaques in senile dementia of the Alzheimer type. Arch Gerontol Geriatr. 1985;4(3):219–29. [DOI] [PubMed] [Google Scholar]
- 155. Hunter JM, Kwan J, Malek‐Ahmadi M, Maarouf CL, Kokjohn TA, Belden C, et al. Morphological and pathological evolution of the brain microcirculation in aging and Alzheimer’s disease. PLoS One. 2012. May;7(5):e36893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Kirabali T, Rust R, Rigotti S, Siccoli A, Nitsch RM, Kulic L. Distinct changes in all major components of the neurovascular unit across different neuropathological stages of Alzheimer’s disease. Brain Pathol. 2020. Aug;30(6):1056–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Kitaguchi H, Ihara M, Saiki H, Takahashi R, Tomimoto H. Capillary beds are decreased in Alzheimer’s disease, but not in Binswanger’s disease. Neurosci Lett. 2007;417(2):128–31. [DOI] [PubMed] [Google Scholar]
- 158. Salat DH, Kaye JA, Janowsky JS. Selective preservation and degeneration within the prefrontal cortex in aging and Alzheimer disease. Arch Neurol. 2001. Sep;58(9):1403. [DOI] [PubMed] [Google Scholar]
- 159. Tisserand DJ, Pruessner JC, Sanz Arigita EJ, van Boxtel MPJ, Evans AC, Jolles J, et al. Regional frontal cortical volumes decrease differentially in aging: an MRI study to compare volumetric approaches and voxel‐based morphometry. NeuroImage. 2002. Oct;17(2):657–69. [PubMed] [Google Scholar]
- 160. Walhovd KB, Fjell AM, Reinvang I, Lundervold A, Dale AM, Eilertsen DE, et al. Effects of age on volumes of cortex, white matter and subcortical structures. Neurobiol Aging. 2005. Oct;26(9):1261–70. [DOI] [PubMed] [Google Scholar]
- 161. Baloyannis SJ, Baloyannis IS. The vascular factor in Alzheimer’s disease: a study in Golgi technique and electron microscopy. J Neurol Sci. 2012;322(1):117–21. [DOI] [PubMed] [Google Scholar]
- 162. Li H‐C, Chen P‐Y, Cheng H‐F, Kuo Y‐M, Huang C‐C. In vivo visualization of brain vasculature in Alzheimer’s disease mice by high‐frequency micro‐doppler imaging. IEEE Trans Biomed Eng. 2019. Dec;66(12). [DOI] [PubMed] [Google Scholar]
- 163. Meyer EP, Ulmann‐Schuler A, Staufenbiel M, Krucker T. Altered morphology and 3D architecture of brain vasculature in a mouse model for Alzheimer’s disease. Proc Natl Acad Sci U S A. 2008. Mar;105(9):3587–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Perlmutter LS, Chui HC, Saperia D, Athanikar J. Microangiopathy and the colocalization of heparan sulfate proteoglycan with amyloid in senile plaques of Alzheimer’s disease. Brain Res. 1990. Jan;508(1):13–9. [DOI] [PubMed] [Google Scholar]
- 165. Zhang X, Yin X, Zhang J, Li A, Gong H, Luo Q, et al. High‐resolution mapping of brain vasculature and its impairment in the hippocampus of Alzheimer’s disease mice. Natl Sci Rev. 2019. Nov;6(6):1223–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Brown WR. A review of string vessels or collapsed, empty basement membrane tubes. J Alzheimers Dis. 2010;21:725–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Ek Olofsson H, Englund E. A cortical microvascular structure in vascular dementia, Alzheimer’s disease, frontotemporal lobar degeneration and nondemented controls: a sign of angiogenesis due to brain ischaemia? Neuropathol Appl Neurobiol. 2019. Oct;45(6):557–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Brown WR, Thore CR. Review: Cerebral microvascular pathology in ageing and neurodegeneration. Neuropathol Appl Neurobiol. 2011. Feb;37(1):56–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Vagnucci AH, Li WW. Alzheimer’s disease and angiogenesis. The Lancet. 2003;361(9357):605–8. [DOI] [PubMed] [Google Scholar]
- 170. Govindpani K, McNamara LG, Smith NR, Vinnakota C, Waldvogel HJ, Faull RLM, et al. Vascular dysfunction in Alzheimer’s disease: a prelude to the pathological process or a consequence of it? J Clin Med. 2019. May;8(5):651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Lau S‐F, Cao H, Fu AKY, Ip NY. Single‐nucleus transcriptome analysis reveals dysregulation of angiogenic endothelial cells and neuroprotective glia in Alzheimer’s disease. Proc Natl Acad Sci U S A. 2020. Oct;117(41):25800–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Grammas P. Neurovascular dysfunction, inflammation and endothelial activation: implications for the pathogenesis of Alzheimer’s disease. J Neuroinflammation. 2011;8(1):26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Alvarez‐Vergara MI, Rosales‐Nieves AE, March‐Diaz R, Rodriguez‐Perinan G, Lara‐Ureña N, Ortega‐de San Luis C, et al. Non‐productive angiogenesis disassembles Aß plaque‐associated blood vessels. Nat Commun. 2021. Dec;12(1):3098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Morawski M, Filippov M, Tzinia A, Tsilibary E, Vargova L. ECM in brain aging and dementia. In: Dityatev A, Wehrle‐Haller B, Pitkänen A, editors. Progress in brain research [Internet]. Elsevier; 2014. p. 207–27. Available from: http://www.sciencedirect.com/science/article/pii/B9780444634863000104 [DOI] [PubMed] [Google Scholar]
- 175. Thomsen MS, Routhe LJ, Moos T. The vascular basement membrane in the healthy and pathological brain. J Cereb Blood Flow Metab. 2017. Jul;37(10):3300–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Zenaro E, Piacentino G, Constantin G. The blood‐brain barrier in Alzheimer’s disease. Neurobiol Dis. 2017. Nov;107:41–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Reed MJ, Damodarasamy M, Banks WA. The extracellular matrix of the blood–brain barrier: structural and functional roles in health, aging, and Alzheimer’s disease. Tissue Barriers. 2019. Oct;7(4):1651157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Ma J, Ma C, Li J, Sun Y, Ye F, Liu K, et al. Extracellular matrix proteins involved in Alzheimer’s disease. Chem Eur J. 2020. Sep;26(53):12101–10. [DOI] [PubMed] [Google Scholar]
- 179. Kurtz A, Oh S‐J. Age related changes of the extracellular matrix and stem cell maintenance. Prev Med. 2012. May;54:S50–6. [DOI] [PubMed] [Google Scholar]
- 180. Hawkes CA, Gatherer M, Sharp MM, Dorr A, Yuen HM, Kalaria R, et al. Regional differences in the morphological and functional effects of aging on cerebral basement membranes and perivascular drainage of amyloid‐β from the mouse brain. Aging Cell. 2013. Apr;12(2):224–36. [DOI] [PubMed] [Google Scholar]
- 181. Scioli M, Bielli A, Arcuri G, Ferlosio A, Orlandi A. Ageing and microvasculature. Vasc Cell. 2014;6(1):19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Zarow C, Barron E, Chui HC, Perlmutter LS. Vascular basement membrane pathology and Alzheimer’s disease. Ann N Y Acad Sci. 1997. Sep;826(1):147–59. [DOI] [PubMed] [Google Scholar]
- 183. Bourasset F, Ouellet M, Tremblay C, Julien C, Do TM, Oddo S, et al. Reduction of the cerebrovascular volume in a transgenic mouse model of Alzheimer’s disease. Neuropharmacology. 2009;56(4):808–13. [DOI] [PubMed] [Google Scholar]
- 184. Merlini M, Meyer EP, Ulmann‐Schuler A, Nitsch RM. Vascular β‐amyloid and early astrocyte alterations impair cerebrovascular function and cerebral metabolism in transgenic arcAβ mice. Acta Neuropathol. 2011. Sep;122(3):293–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Mehta DC, Short JL, Nicolazzo JA. Altered brain uptake of therapeutics in a triple transgenic mouse model of Alzheimer’s disease. Pharm Res. 2013;30(11):2868–79. [DOI] [PubMed] [Google Scholar]
- 186. Lepelletier F‐X, Mann DMA, Robinson AC, Pinteaux E, Boutin H. Early changes in extracellular matrix in Alzheimer’s disease. Neuropathol Appl Neurobiol. 2017. Feb;43(2):167–82. [DOI] [PubMed] [Google Scholar]
- 187. Tong X‐K, Nicolakakis N, Kocharyan A, Hamel E. Vascular remodeling versus amyloid β‐induced oxidative stress in the cerebrovascular dysfunctions associated with Alzheimer’s disease. J Neurosci. 2005. Nov;25(48):11165–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Rockenstein E, Adame A, Mante M, Larrea G, Crews L, Windisch M, et al. Amelioration of the cerebrovascular amyloidosis in a transgenic model of Alzheimer?s disease with the neurotrophic compound Cerebrolysin? J Neural Transm. 2005. Feb;112(2):269–82. [DOI] [PubMed] [Google Scholar]
- 189. Thal DR, Capetillo‐Zarate E, Larionov S, Staufenbiel M, Zurbruegg S, Beckmann N. Capillary cerebral amyloid angiopathy is associated with vessel occlusion and cerebral blood flow disturbances. Neurobiol Aging. 2009;30(12):1936–48. [DOI] [PubMed] [Google Scholar]
- 190. Singh‐Bains MK, Linke V, Austria MDR, Tan AYS, Scotter EL, Mehrabi NF, et al. Altered microglia and neurovasculature in the Alzheimer’s disease cerebellum. Neurobiol Dis. 2019. Dec;132:104589. [DOI] [PubMed] [Google Scholar]
- 191. Christov A, Ottman J, Hamdheydari L, Grammas P. Structural changes in Alzheimers disease brain microvessels. Curr Alzheimer Res. 2008. Aug;5(4):392–5. [DOI] [PubMed] [Google Scholar]
- 192. Donahue JE, Berzin TM, Rafii MS, Glass DJ, Yancopoulos GD, Fallon JR, et al. Agrin in Alzheimer’s disease: altered solubility and abnormal distribution within microvasculature and brain parenchyma. Proc Natl Acad Sci U S A. 1999. May;96(11):6468–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Salloway S, Gur T, Berzin T, Zipser B, Correia S, Hovanesian V, et al. Effect of APOE genotype on microvascular basement membrane in Alzheimer’s disease. J Neurol Sci. 2002. Nov;203–204:183–7. [DOI] [PubMed] [Google Scholar]
- 194. Zipser BD, Johanson CE, Gonzalez L, Berzin TM, Tavares R, Hulette CM, et al. Microvascular injury and blood–brain barrier leakage in Alzheimer’s disease. Neurobiol Aging. 2007;28(7):977–86. [DOI] [PubMed] [Google Scholar]
- 195. Hughes S, Gardiner T, Hu P, Baxter L, Rosinova E, Chan‐Ling T. Altered pericyte–endothelial relations in the rat retina during aging: implications for vessel stability. Neurobiol Aging. 2006. Dec;27(12):1838–47. [DOI] [PubMed] [Google Scholar]
- 196. Hawkes CA, Sullivan PM, Hands S, Weller RO, Nicoll JAR, Carare RO. Disruption of arterial perivascular drainage of amyloid‐β from the brains of mice expressing the human APOE ε4 allele. PLoS One. 2012;7(7):e41636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Farach‐Carson MC, Warren CR, Harrington DA, Carson DD. Border patrol: insights into the unique role of perlecan/heparan sulfate proteoglycan 2 at cell and tissue borders. Matrix Biol. 2014;34:64–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Frantz C, Stewart KM, Weaver VM. The extracellular matrix at a glance. J Cell Sci. 2010. Dec;123(24):4195–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Muenchhoff J, Poljak A, Song F, Raftery M, Brodaty H, Duncan M, et al. Plasma protein profiling of mild cognitive impairment and Alzheimer’s disease across two independent cohorts. J Alzheimers Dis. 2014. Dec;43(4):1355–73. [DOI] [PubMed] [Google Scholar]
- 200. Lemańska‐Perek A, Leszek J, Krzyżanowska‐Gołą;b D, Radzik J, Ką;tnik‐Prastowska I. Molecular status of plasma fibronectin as an additional biomarker for assessment of Alzheimer’s dementia risk. Dement Geriatr Cogn Disord. 2009;28(4):338–42. [DOI] [PubMed] [Google Scholar]
- 201. Murtomäki S, Risteli J, Risteli L, Koivisto U‐M, Johansson S, Liesi P. Laminin and its neurite outgrowth‐promoting domain in the brain in Alzheimer’s disease and Down’s syndrome patients. J Neurosci Res. 1992. Jun;32(2):261–73. [DOI] [PubMed] [Google Scholar]
- 202. Palu E, Liesi P. Differential distribution of laminins in Alzheimer disease and normal human brain tissue. J Neurosci Res. 2002. Jul;69(2):243–56. [DOI] [PubMed] [Google Scholar]
- 203. Matsuda K, Tashiro K, Hayashi Y, Monji A, Yoshida I, Mitsuyama Y. Measurement of laminins in the cerebrospinal fluid obtained from patients with Alzheimer’s disease and vascular dementia using a modified enzyme‐linked immunosorbent assay. Dement Geriatr Cogn Disord. 2002;14(3):113–22. [DOI] [PubMed] [Google Scholar]
- 204. Perlmutter LS, Barrón E, Saperia D, Chui HC. Association between vascular basement membrane components and the lesions of Alzheimer’s disease. J Neurosci Res. 1991. Dec;30(4):673–81. [DOI] [PubMed] [Google Scholar]
- 205. Berzin TM, Zipser BD, Rafii MS, Kuo—Leblanc V, Yancopouloš GD, Glass DJ, et al. Agrin and microvascular damage in Alzheimer’s disease. Neurobiol Aging. 2000. Mar;21(2):349–55. [DOI] [PubMed] [Google Scholar]
- 206. Damodarasamy M, Vernon RB, Pathan JL, Keene CD, Day AJ, Banks WA, et al. The microvascular extracellular matrix in brains with Alzheimer’s disease neuropathologic change (ADNC) and cerebral amyloid angiopathy (CAA). Fluids Barriers CNS. 2020. Sep 29;17(1):60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Carare RO, Bernardes‐Silva M, Newman TA, Page AM, Nicoll JAR, Perry VH, 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. 2008. Apr;34(2):131–44. [DOI] [PubMed] [Google Scholar]
- 208. Morris AWJ, Sharp MM, Albargothy NJ, Fernandes R, Hawkes CA, Verma A, et al. Vascular basement membranes as pathways for the passage of fluid into and out of the brain. Acta Neuropathol. 2016. May;131(5):725–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Rajna Z, Raitamaa L, Tuovinen T, Heikkila J, Kiviniemi V, Seppanen T. 3D multi‐resolution optical flow analysis of cardiovascular pulse propagation in human brain. IEEE Trans Med Imaging. 2019. Sep;38(9):2028–36. [DOI] [PubMed] [Google Scholar]
- 210. 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. 2006. Feb 21;238(4):962–74. [DOI] [PubMed] [Google Scholar]
- 211. Wang P, Olbricht WL. Fluid mechanics in the perivascular space. J Theor Biol. 2011. Apr 7;274(1):52–7. [DOI] [PubMed] [Google Scholar]
- 212. Zekonyte J, Sakai K, Nicoll JAR, Weller RO, Carare RO. Quantification of molecular interactions between ApoE, amyloid‐beta (Aβ) and laminin: relevance to accumulation of Aβ in Alzheimer’s disease. Biochim Biophys Acta Mol Basis Dis. 2016;1862(5):1047–53. [DOI] [PubMed] [Google Scholar]
- 213. Keable A, O’Neill R, MacGregor Sharp M, Gatherer M, Yuen HM, Johnston DA, et al. ApoE4 astrocytes secrete basement membranes rich in fibronectin and poor in laminin compared to ApoE3 astrocytes. Int J Mol Sci. 2020. Jun;21(12):4371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. Conejero‐Goldberg C, Gomar JJ, Bobes‐Bascaran T, Hyde TM, Kleinman JE, Herman MM, et al. APOE2 enhances neuroprotection against Alzheimer’s disease through multiple molecular mechanisms. Mol Psychiatry. 2014. Nov;19(11):1243–50. [DOI] [PubMed] [Google Scholar]
- 215. Kiuchi Y, Isobe Y, Fukushima K. Entactin‐induced inhibition of human amyloid β‐protein fibril formation in vitro. Neurosci Lett. 2001;305(2):119–22. [DOI] [PubMed] [Google Scholar]
- 216. Kiuchi Y, Isobe Y, Fukushima K, Kimura M. Disassembly of amyloid β‐protein fibril by basement membrane components. Life Sci. 2002;70(20):2421–31. [DOI] [PubMed] [Google Scholar]
- 217. Castillo GM, Ngo C, Cummings J, Wight TN, Snow AD. Perlecan binds to the β‐amyloid proteins (Aβ) of Alzheimer’s disease, accelerates Aβ fibril formation, and maintains Aβ fibril stability. J Neurochem. 1997. Dec;69(6):2452–65. [DOI] [PubMed] [Google Scholar]
- 218. Cotman SL, Halfter W, Cole GJ. Agrin binds to β‐amyloid (Aβ), accelerates Aβ fibril formation, and is localized to Aβ deposits in Alzheimer’s disease brain. Mol Cell Neurosci. 2000. Feb;15(2):183–98. [DOI] [PubMed] [Google Scholar]
- 219. Ariga T, Miyatake T, Yu RK. Role of proteoglycans and glycosaminoglycans in the pathogenesis of Alzheimer’s disease and related disorders: amyloidogenesis and therapeutic strategies‐a review. J Neurosci Res. 2010. Aug;88(11):2303–15. [DOI] [PubMed] [Google Scholar]
- 220. Amtul Z, Frías C, Randhawa J, Hill DJ, Arany EJ. The spatial cerebral damage caused by larger infarct and β‐amyloid toxicity is driven by the anatomical/functional connectivity. J Comp Neurol. 2020. Jan;528(1):52–64. [DOI] [PubMed] [Google Scholar]
- 221. Tian J, Shi J, Smallman R, Iwatsubo T, Mann DMA. Relationships in Alzheimer’s disease between the extent of Aβ deposition in cerebral blood vessel walls, as cerebral amyloid angiopathy, and the amount of cerebrovascular smooth muscle cells and collagen. Neuropathol Appl Neurobiol. 2006. Jun;32(3):332–40. [DOI] [PubMed] [Google Scholar]
- 222. Iadecola C. The neurovascular unit coming of age: a journey through neurovascular coupling in health and disease. Neuron. 2017. Sep 27;96(1):17–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Kisler K, Nelson AR, Montagne A, Zlokovic BV. Cerebral blood flow regulation and neurovascular dysfunction in Alzheimer disease. Nat Rev Neurosci. 2017. Jul;18(7):419–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Tamura R, Yoshida K, Toda M. Current understanding of lymphatic vessels in the central nervous system. Neurosurg Rev. 2020. Aug;43(4):1055–64. [DOI] [PubMed] [Google Scholar]
- 225. Valenza M, Facchinetti R, Steardo L, Scuderi C. Altered waste disposal system in aging and Alzheimer’s disease: focus on astrocytic aquaporin‐4. Front Pharmacol. 2019;10:1656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Streit WJ, Khoshbouei H, Bechmann I. The role of microglia in sporadic Alzheimer’s disease. J Alzheimers Dis. 2021;79(3):961–8. [DOI] [PubMed] [Google Scholar]
- 227. Süß P, Schlachetzki JCM. Microglia in Alzheimer’s disease. Curr Alzheimer Res. 2020;17(1):29–43. [DOI] [PubMed] [Google Scholar]
- 228. Donahue JE, Flaherty SL, Johanson CE, Duncan JA, Silverberg GD, Miller MC, et al. RAGE, LRP‐1, and amyloid‐beta protein in Alzheimer’s disease. Acta Neuropathol. 2006. Oct;112(4):405–15. [DOI] [PubMed] [Google Scholar]
- 229. Miller MC, Tavares R, Johanson CE, Hovanesian V, Donahue JE, Gonzalez L, et al. Hippocampal RAGE immunoreactivity in early and advanced Alzheimer’s disease. Brain Res. 2008. Sep;1230:273–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230. Deane R, Du Yan S, Submamaryan RK, LaRue B, Jovanovic S, Hogg E, et al. RAGE mediates amyloid‐beta peptide transport across the blood‐brain barrier and accumulation in brain. Nat Med. 2003. Jul;9(7):907–13. [DOI] [PubMed] [Google Scholar]
- 231. Deane R, Wu Z, Sagare A, Davis J, Du Yan S, Hamm K, et al. LRP/amyloid β‐peptide interaction mediates differential brain efflux of Aβ isoforms. Neuron. 2004. Aug;43(3):333–44. [DOI] [PubMed] [Google Scholar]
- 232. Wu Z, Guo H, Chow N, Sallstrom J, Bell RD, Deane R, et al. Role of the MEOX2 homeobox gene in neurovascular dysfunction in Alzheimer disease. Nat Med. 2005;11(9):959–65. [DOI] [PubMed] [Google Scholar]
- 233. Kalaria RN, Harik SI. Reduced glucose transporter at the blood‐brain barrier and in cerebral cortex in Alzheimer disease. J Neurochem. 1989. Oct;53(4):1083–8. [DOI] [PubMed] [Google Scholar]
- 234. Harik SI. Changes in the glucose transporter of brain capillaries. Can J Physiol Pharmacol. 1992. May;70(S1):S113–7. [DOI] [PubMed] [Google Scholar]
- 235. Simpson IA, Chundu KR, Davies‐Hill T, Honer WG, Davies P. Decreased concentrations of GLUT1 and GLUT3 glucose transporters in the brains of patients with Alzheimer’s disease. Ann Neurol. 1994. May;35(5):546–51. [DOI] [PubMed] [Google Scholar]
- 236. Mooradian AD, Chung HC, Shah GN. GLUT‐1 expression in the cerebra of patients with Alzheimer’s disease. Neurobiol Aging. 1997. Sep;18(5):469–74. [DOI] [PubMed] [Google Scholar]
- 237. Horwood N, Davies DC. Immunolabelling of hippocampal microvessel glucose transporter protein is reduced in Alzheimer’s disease. Virchows Arch. 1994. Aug;425(1):69–72. 10.1007/BF00193951 [DOI] [PubMed] [Google Scholar]
- 238. Vogelsang P, Giil LM, Lund A, Vedeler CA, Parkar AP, Nordrehaug JE, et al. Reduced glucose transporter‐1 in brain derived circulating endothelial cells in mild Alzheimer’s disease patients. Brain Res. 2018. Jan;1678:304–9. [DOI] [PubMed] [Google Scholar]
- 239. Zheng P‐P, Romme E, van der Spek PJ, Dirven CMF, Willemsen R, Kros JM. Glut1/SLC2A1 is crucial for the development of the blood‐brain barrier in vivo. Ann Neurol. 2010. Dec;68(6):835–44. [DOI] [PubMed] [Google Scholar]
- 240. Winkler EA, Nishida Y, Sagare AP, Rege SV, Bell RD, Perlmutter D, et al. GLUT1 reductions exacerbate Alzheimer’s disease vasculo‐neuronal dysfunction and degeneration. Nat Neurosci. 2015. Apr;18(4):521–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241. Kook S‐Y, Hong HS, Moon M, Ha CM, Chang S, Mook‐Jung I. A 1–42‐RAGE interaction disrupts tight junctions of the blood‐brain barrier Via Ca2+‐calcineurin signaling. J Neurosci. 2012. Jun;32(26):8845–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242. Carrano A, Hoozemans JJM, van der Vies SM, Rozemuller AJM, van Horssen J, de Vries HE. Amyloid beta induces oxidative stress‐mediated blood‐brain barrier changes in capillary amyloid angiopathy. Antioxid Redox Signal. 2011. Feb;15(5):1167–78. [DOI] [PubMed] [Google Scholar]
- 243. Carrano A, Hoozemans JJM, van der Vies SM, van Horssen J, de Vries HE, Rozemuller AJM. Neuroinflammation and blood‐brain barrier changes in capillary amyloid angiopathy. Neurodegener Dis. 2012;10(1–4):329–31. [DOI] [PubMed] [Google Scholar]
- 244. Fischer S, Wobben M, Marti HH, Renz D, Schaper W. Hypoxia‐induced hyperpermeability in brain microvessel endothelial cells involves VEGF‐mediated changes in the expression of zonula occludens‐1. Microvasc Res. 2002. Jan;63(1):70–80. [DOI] [PubMed] [Google Scholar]
- 245. Park JH, Okayama N, Gute D, Krsmanovic A, Battarbee H, Alexander JS. Hypoxia/aglycemia increases endothelial permeability: role of second messengers and cytoskeleton. Am J Physiol‐Cell Physiol. 1999. Dec;277(6):C1066–74. [DOI] [PubMed] [Google Scholar]
- 246. Krizbai IA, Bauer H, Bresgen N, Eckl PM, Farkas A, Szatmári E, et al. Effect of oxidative stress on the junctional proteins of cultured cerebral endothelial cells. Cell Mol Neurobiol. 2005. Feb;25(1):129–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247. Fiala M, Liu QN, Sayre J, Pop V, Brahmandam V, Graves MC, et al. Cyclooxygenase‐2‐positive macrophages infiltrate the Alzheimer’s disease brain and damage the blood‐brain barrier. Eur J Clin Invest. 2002. May;32(5):360–71. [DOI] [PubMed] [Google Scholar]
- 248. Biron KE, Dickstein DL, Gopaul R, Jefferies WA. Amyloid triggers extensive cerebral angiogenesis causing blood brain barrier permeability and hypervascularity in Alzheimer’s disease. PLoS One. 2011;6(8):e23789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249. Gauberti M, Fournier AP, Docagne F, Vivien D, Martinez de Lizarrondo S. Molecular magnetic resonance imaging of endothelial activation in the central nervous system. Theranostics. 2018;8(5):1195–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250. Huang C‐W, Tsai M‐H, Chen N‐C, Chen W‐H, Lu Y‐T, Lui C‐C, et al. Clinical significance of circulating vascular cell adhesion molecule‐1 to white matter disintegrity in Alzheimer’s dementia. Thromb Haemost. 2015;114(12):1230–40. [DOI] [PubMed] [Google Scholar]
- 251. Montagne A, Gauberti M, Macrez R, Jullienne A, Briens A, Raynaud J‐S, et al. Ultra‐sensitive molecular MRI of cerebrovascular cell activation enables early detection of chronic central nervous system disorders. NeuroImage. 2012. Nov;63(2):760–70. [DOI] [PubMed] [Google Scholar]
- 252. Drake JD, Chambers AB, Ott BR, Daiello LA; Alzheimer’s Disease Neuroimaging Initiative . Peripheral markers of vascular endothelial dysfunction show independent but additive relationships with brain‐based biomarkers in association with functional impairment in Alzheimer’s disease. J Alzheimers Dis. 2021;80(4):1553–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Zenaro E, Pietronigro E, Bianca VD, Piacentino G, Marongiu L, Budui S, et al. Neutrophils promote Alzheimer’s disease‐like pathology and cognitive decline via LFA‐1 integrin. Nat Med. 2015. Aug;21(8):880–6. [DOI] [PubMed] [Google Scholar]
- 254. Kinney JW, Bemiller SM, Murtishaw AS, Leisgang AM, Salazar AM, Lamb BT. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement. 2018;4:575–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255. Cruz Hernández JC, Bracko O, Kersbergen CJ, Muse V, Haft‐Javaherian M, Berg M, et al. Neutrophil adhesion in brain capillaries reduces cortical blood flow and impairs memory function in Alzheimer’s disease mouse models. Nat Neurosci. 2019. Mar;22(3):413–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256. Winkler EA, Bell RD, Zlokovic BV. Central nervous system pericytes in health and disease. Nat Neurosci. 2011. Oct 26;14(11):1398–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257. Daneman R, Zhou L, Kebede AA, Barres BA. Pericytes are required for blood–brain barrier integrity during embryogenesis. Nature. 2010. Nov;468(7323):562–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258. Armulik A, Abramsson A, Betsholtz C. Endothelial/pericyte interactions. Circ Res. 2005. Sep 16;97(6):512–23. [DOI] [PubMed] [Google Scholar]
- 259. Armulik A, Genové G, Mäe M, Nisancioglu MH, Wallgard E, Niaudet C, et al. Pericytes regulate the blood–brain barrier. Nature. 2010. Nov;468(7323):557–61. [DOI] [PubMed] [Google Scholar]
- 260. Bell RD, Winkler EA, Sagare AP, Singh I, LaRue B, Deane R, et al. Pericytes control key neurovascular functions and neuronal phenotype in the adult brain and during brain aging. Neuron. 2010. Nov;68(3):409–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261. Hall CN, Reynell C, Gesslein B, Hamilton NB, Mishra A, Sutherland BA, et al. Capillary pericytes regulate cerebral blood flow in health and disease. Nature. 2014. Apr;508(7494). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262. Bergers G, Song S. The role of pericytes in blood‐vessel formation and maintenance. Neuro‐Oncol. 2005. Oct 1;7(4):452–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263. Nikolakopoulou AM, Zhao Z, Montagne A, Zlokovic BV. Regional early and progressive loss of brain pericytes but not vascular smooth muscle cells in adult mice with disrupted platelet‐derived growth factor receptor‐β signaling. PLoS One. 2017. Apr;12(4):e0176225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264. Ma Q, Zhao Z, Sagare AP, Wu Y, Wang M, Owens NC, et al. Blood‐brain barrier‐associated pericytes internalize and clear aggregated amyloid‐β42 by LRP1‐dependent apolipoprotein E isoform‐specific mechanism. Mol Neurodegener. 2018. Oct 19;13(1):57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265. Peppiatt CM, Howarth C, Mobbs P, Attwell D. Bidirectional control of CNS capillary diameter by pericytes. Nature. 2006;443(7112):700–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266. Dore‐Duffy P, La Manna JC. Physiologic angiodynamics in the brain. Antioxid Redox Signal. 2007. Jul;9(9):1363–72. [DOI] [PubMed] [Google Scholar]
- 267. Grubb S, Cai C, Hald BO, Khennouf L, Murmu RP, Jensen AGK, et al. Precapillary sphincters maintain perfusion in the cerebral cortex. Nat Commun. 2020. Dec;11(1):395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268. Hamilton NB. Pericyte‐mediated regulation of capillary diameter: a component of neurovascular coupling in health and disease. Front Neuroenergetics. 2010;2:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269. Cai C, Fordsmann JC, Jensen SH, Gesslein B, Lønstrup M, Hald BO, et al. Stimulation‐induced increases in cerebral blood flow and local capillary vasoconstriction depend on conducted vascular responses. Proc Natl Acad Sci U S A. 2018. Jun 19;115(25):E5796–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270. Bonkowski D, Katyshev V, Balabanov RD, Borisov A, Dore‐Duffy P. The CNS microvascular pericyte: pericyte‐astrocyte crosstalk in the regulation of tissue survival. Fluids Barriers CNS. 2011. Jan 18;8(1):8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271. Nakagomi T, Kubo S, Nakano‐Doi A, Sakuma R, Lu S, Narita A, et al. Brain vascular pericytes following ischemia have multipotential stem cell activity to differentiate into neural and vascular lineage cells. Stem Cells. 2015. Jun;33(6):1962–74. [DOI] [PubMed] [Google Scholar]
- 272. Sakuma R, Kawahara M, Nakano‐Doi A, Takahashi AI, Tanaka Y, Narita A, et al. Brain pericytes serve as microglia‐generating multipotent vascular stem cells following ischemic stroke. J Neuroinflammation. 2016. Mar 7;13(1):57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273. Sagare AP, Bell RD, Zhao Z, Ma Q, Winkler EA, Ramanathan A, et al. Pericyte loss influences Alzheimer‐like neurodegeneration in mice. Nat Commun. 2013;4:2932. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 274. Schultz N, Brännström K, Byman E, Moussaud S, Nielsen HM, Olofsson A, et al. Amyloid‐beta 1–40 is associated with alterations in NG2+ pericyte population ex vivo and in vitro. Aging Cell. 2018. Jun;17(3):1–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275. Ding R, Hase Y, Burke M, Foster V, Stevenson W, Polvikoski T, et al. Loss with ageing but preservation of frontal cortical capillary pericytes in post‐stroke dementia, vascular dementia and Alzheimer’s disease. Acta Neuropathol Commun. 2021. Dec;9(1):130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276. Ding R, Hase Y, Ameen‐Ali KE, Ndung'u M, Stevenson W, Barsby J, et al. Loss of capillary pericytes and the blood–brain barrier in white matter in poststroke and vascular dementias and Alzheimer’s disease. Brain Pathol. 2020. Nov;30(6):1087–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277. Miners JS, Schulz I, Love S. Differing associations between Aβ accumulation, hypoperfusion, blood–brain barrier dysfunction and loss of PDGFRB pericyte marker in the precuneus and parietal white matter in Alzheimer’s disease. J Cereb Blood Flow Metab. 2017. Feb;38(1):103–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278. Bourassa P, Tremblay C, Schneider JA, Bennett DA, Calon F. Brain mural cell loss in the parietal cortex in Alzheimer’s disease correlates with cognitive decline and TDP‐43 pathology. Neuropathol Appl Neurobiol. 2020. Aug;46(5):458–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279. Miners JS, Kehoe PG, Love S, Zetterberg H, Blennow K. CSF evidence of pericyte damage in Alzheimer’s disease is associated with markers of blood‐brain barrier dysfunction and disease pathology. Alzheimers Res Ther. 2019. Dec;11(1):81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280. Halliday MR, Rege SV, Ma Q, Zhao Z, Miller CA, Winkler EA, et al. Accelerated pericyte degeneration and blood–brain barrier breakdown in apolipoprotein E4 carriers with Alzheimer’s disease. J Cereb Blood Flow Metab. 2016. Jan;36(1):216–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281. Sun X, He G, Qing H, Zhou W, Dobie F, Cai F, et al. Hypoxia facilitates Alzheimer’s disease pathogenesis by up‐regulating BACE1 gene expression. Proc Natl Acad Sci U S A. 2006. Dec 5;103(49):18727–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282. Zhang X, Zhou K, Wang R, Cui J, Lipton SA, Liao F‐F, et al. Hypoxia‐inducible factor 1α (HIF‐1α)‐mediated hypoxia increases BACE1 expression and β‐amyloid generation. J Biol Chem. 2007. Apr;282(15):10873–80. [DOI] [PubMed] [Google Scholar]
- 283. Hibbs E, Love S, Miners JS. Pericyte contractile responses to endothelin‐1 and aβ peptides: assessment by electrical impedance assay. Front Cell Neurosci. 2021;15:723953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284. Sharif Y, Jumah F, Coplan L, Krosser A, Sharif K, Tubbs RS. Blood brain barrier: a review of its anatomy and physiology in health and disease: blood brain barrier in health and disease. Clin Anat. 2018. Sep;31(6):812–23. [DOI] [PubMed] [Google Scholar]
- 285. Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV. Blood‐brain barrier: from physiology to disease and back. Physiol Rev. 2019. Jan 1;99(1):21–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286. Serlin Y, Shelef I, Knyazer B, Friedman A. Anatomy and physiology of the blood–brain barrier. Semin Cell Dev Biol. 2015. Feb;38:2–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287. Pandit R, Chen L, Götz J. The blood‐brain barrier: physiology and strategies for drug delivery. Adv Drug Deliv Rev. 2020;165–166:1–14. [DOI] [PubMed] [Google Scholar]
- 288. Liebner S, Dijkhuizen RM, Reiss Y, Plate KH, Agalliu D, Constantin G. Functional morphology of the blood–brain barrier in health and disease. Acta Neuropathol. 2018. Mar;135(3):311–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289. Yamazaki Y, Kanekiyo T. Blood‐brain barrier dysfunction and the pathogenesis of Alzheimer’s disease. Int J Mol Sci. 2017. Sep;18(9):1965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290. Chakraborty A, Chatterjee M, Twaalfhoven H, Del Campo Milan M, Teunissen CE, Scheltens P, et al. Vascular endothelial growth factor remains unchanged in cerebrospinal fluid of patients with Alzheimer’s disease and vascular dementia. Alzheimers Res Ther. 2018. Dec;10(1):58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291. Cai Z, Qiao P‐F, Wan C‐Q, Cai M, Zhou N‐K, Li Q. Role of blood‐brain barrier in Alzheimer’s disease. J Alzheimers Dis. 2018. May;63(4):1223–34. [DOI] [PubMed] [Google Scholar]
- 292. Montagne A, Zhao Z, Zlokovic BV. Alzheimer’s disease: a matter of blood–brain barrier dysfunction? J Exp Med. 2017. Nov;214(11):3151–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293. Blair LJ, Frauen HD, Zhang BO, Nordhues BA, Bijan S, Lin Y‐C, et al. Tau depletion prevents progressive blood‐brain barrier damage in a mouse model of tauopathy. Acta Neuropathol Commun. 2015. Dec;3(1):8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294. Wisniewski HM, Kozlowski PB. Evidence for blood‐brain barrier changes in senile dementia of the Alzheimer type (SDAT). Ann N Y Acad Sci. 1982;396:119–29. [DOI] [PubMed] [Google Scholar]
- 295. Ryu JK, McLarnon JG. A leaky blood‐brain barrier, fibrinogen infiltration and microglial reactivity in inflamed Alzheimer’s disease brain. J Cell Mol Med. 2009. Sep;13(9A):2911–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296. Cortes‐Canteli M, Mattei L, Richards AT, Norris EH, Strickland S. Fibrin deposited in the Alzheimer’s disease brain promotes neuronal degeneration. Neurobiol Aging. 2015;36(2):608–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297. Hultman K, Strickland S, Norris EH. The APOE ε4/ε4 genotype potentiates vascular fibrin(ogen) deposition in amyloid‐laden vessels in the brains of Alzheimer’s disease patients. J Cereb Blood Flow Metab. 2013. Aug;33(8):1251–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298. Halliday MR, Pomara N, Sagare AP, Mack WJ, Frangione B, Zlokovic BV. Relationship between cyclophilin a levels and matrix metalloproteinase 9 Activity in cerebrospinal fluid of cognitively normal apolipoprotein E4 carriers and blood‐brain barrier breakdown. JAMA Neurol. 2013. Sep;70(9):1198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299. Skoog I, Wallin A, Fredman P, Hesse C, Aevarsson O, Karlsson I, et al. A population study on blood‐brain barrier function in 85‐year‐olds: relation to Alzheimer’s disease and vascular dementia. Neurology. 1998. Apr;50(4):966–71. [DOI] [PubMed] [Google Scholar]
- 300. Janelidze S, Hertze J, Nägga K, Nilsson K, Nilsson C, Wennström M, et al. Increased blood‐brain barrier permeability is associated with dementia and diabetes but not amyloid pathology or APOE genotype. Neurobiol Aging. 2017. Mar;51:104–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301. Skillbäck T, Delsing L, Synnergren J, Mattsson N, Janelidze S, Nägga K, et al. CSF/serum albumin ratio in dementias: a cross‐sectional study on 1861 patients. Neurobiol Aging. 2017. Nov;59:1–9. [DOI] [PubMed] [Google Scholar]
- 302. Baker SK, Chen Z‐L, Norris EH, Revenko AS, MacLeod AR, Strickland S. Blood‐derived plasminogen drives brain inflammation and plaque deposition in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci U S A. 2018. Oct;115(41):E9687–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303. Montagne A, Nation DA, Pa J, Sweeney MD, Toga AW, Zlokovic BV. Brain imaging of neurovascular dysfunction in Alzheimer’s disease. Acta Neuropathol. 2016. May;131(5):687–707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304. van de Haar HJ, Burgmans S, Jansen JFA, van Osch MJP, van Buchem MA, Muller M, et al. Blood‐brain barrier leakage in patients with early Alzheimer disease. Radiology. 2016. Nov;281(2):527–35. [DOI] [PubMed] [Google Scholar]
- 305. van de Haar HJ, Jansen JFA, Jeukens CRLPN, Burgmans S, van Buchem MA, Muller M, et al. Subtle blood‐brain barrier leakage rate and spatial extent: considerations for dynamic contrast‐enhanced MRI. Med Phys. 2017. Aug;44(8):4112–25. [DOI] [PubMed] [Google Scholar]
- 306. Montagne A, Nation DA, Sagare AP, Barisano G, Sweeney MD, Chakhoyan A, et al. APOE4 leads to blood–brain barrier dysfunction predicting cognitive decline. Nature. 2020. May;581(7806):71–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 307. Yang Y, Rosenberg GA. Blood‐brain barrier breakdown in acute and chronic cerebrovascular disease. Stroke. 2011. Nov;42(11):3323–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 308. Asahi M, Asahi K, Jung JC, del Zoppo GJ, Fini ME, Lo EH. Role for matrix metalloproteinase 9 after focal cerebral ischemia: effects of gene knockout and enzyme inhibition with BB‐94. J Cereb Blood Flow Metab. 2000. Dec;20(12):1681–9. [DOI] [PubMed] [Google Scholar]
- 309. Brkic M, Balusu S, Van Wonterghem E, Gorle N, Benilova I, Kremer A, et al. Amyloid oligomers disrupt blood‐CSF barrier integrity by activating matrix metalloproteinases. J Neurosci. 2015. Sep;35(37):12766–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 310. Takata F, Dohgu S, Matsumoto J, Takahashi H, Machida T, Wakigawa T, et al. Brain pericytes among cells constituting the blood‐brain barrier are highly sensitive to tumor necrosis factor‐α, releasing matrix metalloproteinase‐9 and migrating in vitro. J Neuroinflammation. 2011;8(1):106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 311. Torres‐Acosta N, O’Keefe JH, O’Keefe EL, Isaacson R, Small G. Therapeutic potential of TNF‐α inhibition for Alzheimer’s disease prevention. J Alzheimers Dis. 2020;78(2):619–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 312. Haas TL. Endothelial cell regulation of matrix metalloproteinases. Can J Physiol Pharmacol. 2005. Jan 1;83(1):1–7. [DOI] [PubMed] [Google Scholar]
- 313. Milkiewicz M, Ispanovic E, Doyle JL, Haas TL. Regulators of angiogenesis and strategies for their therapeutic manipulation. Int J Biochem Cell Biol. 2006. Mar;38(3):333–57. [DOI] [PubMed] [Google Scholar]
- 314. Hoffmann S, He S, Ehren M, Ryan SJ, Wiedemann P, Hinton DR. mmp‐2 and mmp‐9 secretion by rpe is stimulated by angiogenic molecules found in choroidal neovascular membranes. Retina. 2006. Apr;26(4):454–61. [DOI] [PubMed] [Google Scholar]
- 315. Shi H, Koronyo Y, Rentsendorj A, Fuchs D‐T, Sheyn J, Black KL, et al. Retinal vasculopathy in Alzheimer’s disease. Front Neurosci. 2021. Sep 22;15:731614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 316. Shi H, Koronyo Y, Rentsendorj A, Regis GC, Sheyn J, Fuchs D‐T, et al. Identification of early pericyte loss and vascular amyloidosis in Alzheimer’s disease retina. Acta Neuropathol. 2020. May;139(5):813–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317. Vandenabeele M, Veys L, Lemmens S, Hadoux X, Gelders G, Masin L, et al. The AppNL‐G‐F mouse retina is a site for preclinical Alzheimer’s disease diagnosis and research. Acta Neuropathol Commun. 2021. Dec;9(1):6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 318. Koustenis A, Harris A, Gross J, Januleviciene I, Shah A, Siesky B. Optical coherence tomography angiography: an overview of the technology and an assessment of applications for clinical research. Br J Ophthalmol. 2017. Jan;101(1):16–20. [DOI] [PubMed] [Google Scholar]
- 319. Zhang YS, Zhou N, Knoll BM, Samra S, Ward MR, Weintraub S, et al. Parafoveal vessel loss and correlation between peripapillary vessel density and cognitive performance in amnestic mild cognitive impairment and early Alzheimer’s Disease on optical coherence tomography angiography. PLoS One. 2019. Apr;14(4):e0214685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 320. Yoon SP, Grewal DS, Thompson AC, Polascik BW, Dunn C, Burke JR, et al. Retinal microvascular and neurodegenerative changes in Alzheimer’s disease and mild cognitive impairment compared with control participants. Ophthalmol Retina. 2019;3(6):489–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 321. Jiang H, Wei Y, Shi Y, Wright CB, Sun X, Gregori G, et al. Altered macular microvasculature in mild cognitive impairment and Alzheimer disease. J Neuroophthalmol. 2018;38(3):292–8. 10.1097/WNO.0000000000000580 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 322. Grewal DS, Polascik BW, Hoffmeyer GC, Fekrat S. Assessment of differences in retinal microvasculature using OCT angiography in Alzheimer’s disease: a twin discordance report. Ophthalmic Surg Lasers Imaging Retina. 2018. Jun;49(6):440–4. [DOI] [PubMed] [Google Scholar]
- 323. Bulut M, Kurtuluş F, Gözkaya O, Erol MK, Cengiz A, Akıdan M, et al. Evaluation of optical coherence tomography angiographic findings in Alzheimer’s type dementia. Br J Ophthalmol. 2018. Feb;102(2):233–7. [DOI] [PubMed] [Google Scholar]
- 324. Yan Y, Wu X, Wang X, Geng Z, Wang L, Xiao G, et al. The retinal vessel density can reflect cognitive function in patients with Alzheimer’s disease: evidence from optical coherence tomography angiography. J Alzheimers Dis. 2021. Jan 5;1–10. [DOI] [PubMed] [Google Scholar]
- 325. Wang XI, Zhao Q, Tao R, Lu H, Xiao Z, Zheng LI, et al. Decreased retinal vascular density in Alzheimer’s disease (AD) and mild cognitive impairment (MCI): an optical coherence tomography angiography (OCTA) study. Front Aging Neurosci. 2021. Jan 15;12:572484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 326. Chua J, Hu Q, Ke M, Tan B, Hong J, Yao X, et al. Retinal microvasculature dysfunction is associated with Alzheimer’s disease and mild cognitive impairment. Alzheimers Res Ther. 2020. Dec 4;12(1):161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 327. Wu J, Zhang X, Azhati G, Li T, Xu G, Liu F. Retinal microvascular attenuation in mental cognitive impairment and Alzheimer’s disease by optical coherence tomography angiography. Acta Ophthalmol. 2020. Sep;98(6):e781–7. 10.1111/aos.14381 [DOI] [PubMed] [Google Scholar]
- 328. O’Bryhim BE, Apte RS, Kung N, Coble D, Van Stavern GP. Association of preclinical Alzheimer disease with optical coherence tomographic angiography findings. JAMA Ophthalmol. 2018. Nov;136(11):1242–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 329. Haan J, Kreeke JA, Berckel BN, Barkhof F, Teunissen CE, Scheltens P, et al. Is retinal vasculature a biomarker in amyloid proven Alzheimer’s disease? Alzheimers Dement Diagn Assess Dis Monit. 2019;11:383–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 330. Salobrar‐Garcia E, Méndez‐Hernández C, Hoz RD, Ramírez AI, López‐Cuenca I, Fernández‐Albarral JA, et al. Ocular vascular changes in mild Alzheimer’s disease patients: foveal avascular zone. choroidal thickness, and ONH hemoglobin analysis. J Pers Med. 2020. Nov 15;10(4):231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 331. Shin JY, Choi EY, Kim M, Lee HK, Byeon SH. Changes in retinal microvasculature and retinal layer thickness in association with apolipoprotein E genotype in Alzheimer’s disease. Sci Rep. 2021. Dec;11(1):1847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 332. van de Kreeke JA, Nguyen H‐T, Konijnenberg E, Tomassen J, den Braber A, ten Kate M, et al. Optical coherence tomography angiography in preclinical Alzheimer’s disease. Br J Ophthalmol. 2020. Feb;104(2):157–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 333. Czakó C, Kovács T, Ungvari Z, Csiszar A, Yabluchanskiy A, Conley S, et al. Retinal biomarkers for Alzheimer’s disease and vascular cognitive impairment and dementia (VCID): implication for early diagnosis and prognosis. GeroScience. 2020. Dec;42(6):1499–525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 334. Song A, Johnson N, Ayala A, Thompson AC. Optical coherence tomography in patients with Alzheimer’s disease: what can it tell us? Eye Brain. 2021;13:1–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 335. Lee CS, Apte RS. Retinal biomarkers of Alzheimer disease. Am J Ophthalmol. 2020. Oct;218:337–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 336. Alber J, Goldfarb D, Thompson LI, Arthur E, Hernandez K, Cheng D, et al. Developing retinal biomarkers for the earliest stages of Alzheimer’s disease: what we know, what we don’t, and how to move forward. Alzheimers Dement. 2020. Jan;16(1):229–43. [DOI] [PubMed] [Google Scholar]
- 337. Chew G, Petretto E. Transcriptional networks of microglia in Alzheimer’s disease and insights into pathogenesis. Genes. 2019. Oct 12;10(10):798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 338. Schwabe T, Srinivasan K, Rhinn H. Shifting paradigms: the central role of microglia in Alzheimer’s disease. Neurobiol Dis. 2020. Sep;143:104962. [DOI] [PubMed] [Google Scholar]
- 339. Hashemiaghdam A, Mroczek M. Microglia heterogeneity and neurodegeneration: the emerging paradigm of the role of immunity in Alzheimer’s disease. J Neuroimmunol. 2020. Apr;341:577185. [DOI] [PubMed] [Google Scholar]
- 340. Gerrits E, Heng Y, Boddeke EWGM, Eggen BJL. Transcriptional profiling of microglia; current state of the art and future perspectives. Glia. 2020. Apr;68(4):740–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 341. Mathys H, Davila‐Velderrain J, Peng Z, Gao F, Mohammadi S, Young JZ, et al. Single‐cell transcriptomic analysis of Alzheimer’s disease. Nature. 2019. Jun;570(7761):332–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 342. Zhou Y, Song WM, Andhey PS, Swain A, Levy T, Miller KR, et al. Human and mouse single‐nucleus transcriptomics reveal TREM2‐dependent and TREM2‐independent cellular responses in Alzheimer’s disease. Nat Med. 2020. Jan;26(1):131–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 343. Grubman A, Chew G, Ouyang JF, Sun G, Choo XY, McLean C, et al. A single‐cell atlas of entorhinal cortex from individuals with Alzheimer’s disease reveals cell‐type‐specific gene expression regulation. Nat Neurosci. 2019. Dec;22(12):2087–97. [DOI] [PubMed] [Google Scholar]
- 344. Song HW, Foreman KL, Gastfriend BD, Kuo JS, Palecek SP, Shusta EV. Transcriptomic comparison of human and mouse brain microvessels. Sci Rep. 2020. Dec;10(1):12358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 345. Yang AC, Vest RT, Kern F, Lee DP, Agam M, Maat CA, et al. A human brain vascular atlas reveals diverse mediators of Alzheimer’s risk. Nature. 2022. 10.1038/s41586-021-04369-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 346. Wang Z, Zhang Q, Lin J‐R, Jabalameli MR, Mitra J, Nguyen N, et al. Deep post‐GWAS analysis identifies potential risk genes and risk variants for Alzheimer’s disease, providing new insights into its disease mechanisms. Sci Rep. 2021. Oct 15;11(1):20511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 347. Lake BB, Chen S, Sos BC, Fan J, Kaeser GE, Yung YC, et al. Integrative single‐cell analysis of transcriptional and epigenetic states in the human adult brain. Nat Biotechnol. 2018. Jan;36(1):70–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 348. Chakraborty A, de Wit NM, van der Flier WM, de Vries HE. The blood brain barrier in Alzheimer’s disease. Vascul Pharmacol. 2017;89:12–8. [DOI] [PubMed] [Google Scholar]
- 349. Delafontaine‐Martel P, Lefebvre J, Damseh R, Castonguay A, Tardif P, Lesage F. Large scale serial two‐photon microscopy to investigate local vascular changes in whole rodent brain models of Alzheimer’s disease. Multiphoton Microscopy Biomed Sci XVIII. 2018;10498:104982O. 10.1117/12.2290060 [DOI] [Google Scholar]
- 350. Zhang L‐Y, Lin P, Pan J, Ma Y, Wei Z, Jiang LU, et al. CLARITY for high‐resolution imaging and quantification of vasculature in the whole mouse brain. Aging Dis. 2018;9(2):262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 351. Di Giovanna AP, Tibo A, Silvestri L, Müllenbroich MC, Costantini I, Allegra Mascaro AL, et al. Whole‐brain vasculature reconstruction at the single capillary level. Sci Rep. 2018. Aug 22;8(1):12573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 352. Tong L, Hill RA, Damisah EC, Murray KN, Yuan P, Bordey A, et al. Imaging and optogenetic modulation of vascular mural cells in the live brain. Nat Protoc. 2021. Jan;16(1):472–96. [DOI] [PubMed] [Google Scholar]
- 353. Shabir O, Sharp P, Rebollar MA, Boorman L, Howarth C, Wharton SB, et al. Enhanced cerebral blood volume under normobaric hyperoxia in the J20‐hAPP mouse model of Alzheimer’s disease. Sci Rep. 2020. May 5;10(1):7518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 354. Bell MA, Ball MJ. The correlation of vascular capacity with the parenchymal lesions of Alzheimer’s disease. Can J Neurol Sci. 1986. Nov;13(S4):456–61. [DOI] [PubMed] [Google Scholar]
- 355. Kawai M, Kalaria RN, Harik SI, Perry G. The relationship of amyloid plaques to cerebral capillaries in Alzheimer’s disease. Am J Pathol. 1990. Dec;137(6):1435–46. [PMC free article] [PubMed] [Google Scholar]
- 356. Bell MA, Ball MJ. Neuritic plaques and vessels of visual cortex in aging and Alzheimer’s dementia. Neurobiol Aging. 1990;11(4):359–70. [DOI] [PubMed] [Google Scholar]
- 357. Harris R, Miners JS, Allen S, Love S. VEGFR1 and VEGFR2 in Alzheimer’s disease. J Alzheimers Dis. 2018;61:741–52. [DOI] [PubMed] [Google Scholar]
- 358. Thomas T, Miners S, Love S. Post‐mortem assessment of hypoperfusion of cerebral cortex in Alzheimer’s disease and vascular dementia. Brain. 2015. Feb;138(4):1059–69. [DOI] [PubMed] [Google Scholar]
- 359. Brown WR, Moody DM, Thore CR, Challa VR, Anstrom JA. Vascular dementia in leukoaraiosis may be a consequence of capillary loss not only in the lesions, but in normal‐appearing white matter and cortex as well. J Neurol Sci. 2007. Jun;257(1–2):62–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 360. Buée L, Hof PR, Bouras C, Delacourte A, Perl DP, Morrison JH, et al. Pathological alterations of the cerebral microvasculature in Alzheimer’s disease and related dementing disorders. Acta Neuropathol. 1994;87(5):469–80. [DOI] [PubMed] [Google Scholar]
- 361. Nikolajsen GN, Kotynski KA, Jensen MS, West MJ. Quantitative analysis of the capillary network of aged APPswe/PS1dE9 transgenic mice. Neurobiol Aging. 2015. Nov;36(11):2954–62. [DOI] [PubMed] [Google Scholar]
- 362. Paris D, Patel N, DelleDonne A, Quadros A, Smeed R, Mullan M. Impaired angiogenesis in a transgenic mouse model of cerebral amyloidosis. Neurosci Lett. 2004;366(1):80–5. [DOI] [PubMed] [Google Scholar]
- 363. Herring A, Münster Y, Metzdorf J, Bolczek B, Krüssel S, Krieter D, et al. Late running is not too late against Alzheimer’s pathology. Neurobiol Dis. 2016;94:44–54. [DOI] [PubMed] [Google Scholar]
- 364. Zhang H, Jin B, Faber JE. Mouse models of Alzheimer’s disease cause rarefaction of pial collaterals and increased severity of ischemic stroke. Angiogenesis. 2019;22(2):263–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 365. Miao J, Xu F, Davis J, Otte‐Höller I, Verbeek MM, Van Nostrand WE. Cerebral microvascular amyloid beta protein deposition induces vascular degeneration and neuroinflammation in transgenic mice expressing human vasculotropic mutant amyloid beta precursor protein. Am J Pathol. 2005. Aug;167(2):505–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 366. Keyvani K, Münster Y, Kurapati NK, Rubach S, Schönborn A, Kocakavuk E, et al. Higher levels of kallikrein‐8 in female brain may increase the risk for Alzheimer’s disease. Brain Pathol. 2018. Nov;28(6):947–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 367. Lee GD, Aruna JH, Barrett PM, Lei D‐L, Ingram DK, Mouton PR. Stereological analysis of microvascular parameters in a double transgenic model of Alzheimer’s disease. Brain Res Bull. 2005;65(4):317–22. [DOI] [PubMed] [Google Scholar]
- 368. Kouznetsova E, Klingner M, Sorger D, Sabri O, Großmann U, Steinbach J, et al. Developmental and amyloid plaque‐related changes in cerebral cortical capillaries in transgenic Tg2576 Alzheimer mice. Int J Dev Neurosci. 2006. Apr;24(2–3):187–93. [DOI] [PubMed] [Google Scholar]
- 369. Stefanova NA, Maksimova KY, Rudnitskaya EA, Muraleva NA, Kolosova NG. Association of cerebrovascular dysfunction with the development of Alzheimer’s disease‐like pathology in OXYS rats. BMC Genom. 2018;19(3):75. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Not applicable.