Next Article in Journal
Genome-Wide Characterization, Evolution, and Expression Profile Analysis of GATA Transcription Factors in Brachypodium distachyon
Next Article in Special Issue
Plasma Amyloid-Beta Levels in a Pre-Symptomatic Dutch-Type Hereditary Cerebral Amyloid Angiopathy Pedigree: A Cross-Sectional and Longitudinal Investigation
Previous Article in Journal
Exosomal Cargo May Hold the Key to Improving Reproductive Outcomes in Dairy Cows
Previous Article in Special Issue
A Toxic Synergy between Aluminium and Amyloid Beta in Yeast
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Neurovascular Unit Dysfunction in Alzheimer’s Disease

by
Luis O. Soto-Rojas
1,*,
Mar Pacheco-Herrero
2,*,
Paola A. Martínez-Gómez
1,
B. Berenice Campa-Córdoba
3,4,
Ricardo Apátiga-Pérez
3,4,
Marcos M. Villegas-Rojas
5,
Charles R. Harrington
6,
Fidel de la Cruz
3,
Linda Garcés-Ramírez
3 and
José Luna-Muñoz
4,7,*
1
Facultad de Estudios Superiores Iztacala, UNAM, Mexico City 54090, Mexico
2
Neuroscience Research Laboratory, Faculty of Health Sciences, Pontificia Universidad Catolica Madre y Maestra, Santiago de los Caballeros 51000, Dominican Republic
3
Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07738, Mexico
4
National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores, Cuautitlán, UNAM, Mexico City 53150, Mexico
5
Unidad Profesional Interdisciplinaria de Biotecnología del Instituto Politécnico Nacional (UPIBI-IPN), Mexico City 07340, Mexico
6
School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
7
Banco Nacional de Cerebros-UNPHU, Universidad Nacional Pedro Henríquez Ureña, Santo Domingo 2796, Dominican Republic
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(4), 2022; https://doi.org/10.3390/ijms22042022
Submission received: 16 December 2020 / Revised: 6 January 2021 / Accepted: 11 January 2021 / Published: 18 February 2021
(This article belongs to the Special Issue Molecular Mechanism of Alzheimer's Disease III)

Abstract

:
Alzheimer’s disease (AD) is the most common neurodegenerative disease worldwide. Histopathologically, AD presents with two hallmarks: neurofibrillary tangles (NFTs), and aggregates of amyloid β peptide (Aβ) both in the brain parenchyma as neuritic plaques, and around blood vessels as cerebral amyloid angiopathy (CAA). According to the vascular hypothesis of AD, vascular risk factors can result in dysregulation of the neurovascular unit (NVU) and hypoxia. Hypoxia may reduce Aβ clearance from the brain and increase its production, leading to both parenchymal and vascular accumulation of Aβ. An increase in Aβ amplifies neuronal dysfunction, NFT formation, and accelerates neurodegeneration, resulting in dementia. In recent decades, therapeutic approaches have attempted to decrease the levels of abnormal Aβ or tau levels in the AD brain. However, several of these approaches have either been associated with an inappropriate immune response triggering inflammation, or have failed to improve cognition. Here, we review the pathogenesis and potential therapeutic targets associated with dysfunction of the NVU in AD.

1. Introduction

The neurovascular unit (NVU) is defined as a complex functional and anatomical structure composed of: (a) neurons and interneurons, (b) glial cells such as microglia, astrocytes, and oligodendrocytes, (c) vascular cells such as endothelial cells, pericytes, and smooth muscle cells (SMCs), and (d) a basal lamina formed by brain endothelial cells and extracellular matrix (Figure 1) [1,2]. The NVU components are intimately linked to each other, enabling an efficient system for cerebral blood flow (CBF), maintenance of neuronal metabolic activity [3], and an effective blood-brain barrier (BBB). The BBB is a specialized structure in the cerebral vasculature formed by astrocytes, pericytes, and specialized junctions such as tight-junctions (TJs) and adherent-junctions (AJs) of endothelial cells [4]. The BBB serves to limit the entry of pathogens, toxic agents, and blood cells into the brain parenchyma [5]. The junctional components of the NVU are made by gap junctions and adhesion molecules such as integrins and cadherins [6]. This interplay facilitates the influx/efflux of Ca2+, K+, and the neuromodulatory action of ATP [7]. Each NVU component plays an active and specific role in maintaining the dynamic linkages reciprocally under physiological conditions [8]. Dysregulation of the NVU and BBB are critical pathophysiological events in neurodegenerative diseases, including Alzheimer’s disease (AD) [9].
AD is one of the most important neurodegenerative disorders worldwide and the leading cause of cognitive and functional decline in the elderly [10]. The classic neuropathological hallmarks of AD involve aggregates of tau and amyloid β-peptides (Aβ). Neurofibrillary tangles (NFTs) and hyperphosphorylated tau accumulate intracellularly and are typically accompanied by neuronal loss [11,12]. The second hallmark is the presence of amyloid β-peptide (Aβ) deposits in the brain parenchyma and around cerebral blood vessels as neuritic plaques (NPs) and cerebral amyloid angiopathy (CAA), respectively. CAA is characterized by Aβ deposition within the walls of cortical and leptomeningeal arteries and veins [13], which lead to NVU dysfunction [11,12]. CAA is universally found in AD brains [14] and has been linked to neuroinflammation, chronic hypoperfusion, ischemia, and loss of the blood vessel wall integrity and hemorrhage [15,16].
In this study, we review the relationship between the NVU and BBB dysregulation and AD pathology. Furthermore, possible therapeutic targets aimed at preventing or restoring NVU in AD are discussed.

2. Cellular and Structural Components of the NVU Along the Cerebrovascular Tree

An extensive system of arteries, arterioles, and capillaries, delivers oxygenated blood and nutrients to the brain. The pial arteries derived from arteries go along the brain surface and penetrate the parenchyma, branching into arterioles and capillaries [17]. The cellular NVU composition changes along the cerebrovascular tree (Figure 1a):
(a)
Pial arteries consist of multiple layers of SMCs, separated from the endothelium by a notable elastic lamina (Figure 1b), and innervated by nerve fibers formed from sensory and peripheral autonomic ganglia [18]. The pial arteries penetrating the brain are surrounded by the subarachnoid space (SAS; Figure 1b). Traditionally, the pial arteries have been associated with CBF and neuronal homeostasis [19].
(b)
Penetrating arterioles have several thin SMC layers that become a single layer (Figure 1c) [20]. The density of perivascular nerves is low at this level, and the elastic lamina becomes less prominent [20]. The perivascular space (also known as “Virchow-Robin space”) is delimited by the astrocytic end-foot (glia limitans) and the vascular basement membrane [21]. It comprises different cell types, including perivascular macrophages (PVMs), pial cells, Mato cells and mast cells, and collagen and nerve fibers [21] (Figure 1c). The perivascular space is an exchange pathway for the glymphatic system. The glymphatic system has been defined as a network of perivascular pathways that favors the exchange of both solutes and liquids between the cerebrospinal fluid (CSF) and the interstitial compartments, promoting clearance of metabolites, proteins, and debris from the brain interstitium [22,23,24]. This clearance depends mainly on the aquaporin-4 water channels (AQP4), found in astrocytic end-feet [25]. It has been demonstrated that the pial and penetrating arterioles can regulate arterial tone by the extrinsic and intrinsic innervations, respectively [26].
(c)
The intraparenchymal arterioles are formed when the arterioles invade deeper into the brain. Here, the glial limitans and the vascular basement membrane fuse, eliminating the Virchow-Robin space [21]. These arterioles have a single layer of SMCs, lack perivascular nerves, and are encapsulated by the astrocytic end-feet (Figure 1d) [20]. Endothelial cells extend their protrusions to SMCs and connect through gap junctions [27]. They are related to functional hyperemia, which ensures a rapid increase in the CBF rate to activated brain structures [28].
(d)
Capillaries are the smallest vessels in the brain and exchange molecules between blood and brain across the BBB [29]. Pericytes replace the SMCs, and mural cells are immersed in the endothelial basement membrane (Figure 1e) [30]. The border of the capillaries is enveloped by astrocytic end-feet, and the neural processes can be adjacent to the capillary basal lamina (Figure 1e) [31]. Pericytes and endothelial cells make direct interdigitated contacts where cytoplasmic protrusions (pegs) of one cell type insert into the opposing cell membrane (socket) of the other cell type [32].

3. The Physiological Characteristics of the Blood-Brain Barrier (BBB)

The endothelial cells that make up the brain capillaries are tightly sealed and act as a barrier, referred to as the “blood-brain barrier” (BBB). The BBB is a specialized brain endothelial composition of the neurovascular system that is completely differentiated. Together with astrocytic end-feet, pericytes, and microglia, the BBB separates the circulating blood components from neurons [2,33] (Figure 1e and Figure 2a).
The nearness of the different cell types with one another in the NVU allows for an effective paracrine regulation, the maintenance of the BBB, and the normal functioning of the central nervous system (CNS), such as synaptic transmission and remodeling, neurogenesis, and angiogenesis in the adult brain [2]. The cell-to-cell hermetic contact confers to the BBB the properties of low paracellular and transcellular permeability and high transendothelial electrical resistance [29].
The firmly sealed endothelium limits the entry of most blood-derived particles into the brain unless they possess receptors or carriers (glucose, hormones, amino acids, and nucleotides) to be transported across the BBB [34]. The concentration gradient is the most considerable factor for carrier-mediated transport across the BBB and is preferentially influenced by the affinity, size, and physiochemical properties of each specific molecule [35]. The BBB excludes typically free exchange of solutes between blood-brain and brain-blood [36]. One exception is that of small lipid-soluble molecules <400 Da, which can cross the BBB via lipid-mediated diffusion [37], adsorptive endocytosis, and receptor-mediated endocytosis [38]. On the other hand, transendothelial passive diffusion enables the small influx of lipophilic and nonpolar molecules into brains across the lipid bilayer of endothelial cells [38].
Endothelial cells are typically connected at a junctional complex by the tight-junctions (TJs) and adherent-junctions (AJs) (Figure 2a) [39]. The TJs provide low paracellular permeability, high electrical resistance [40], and binding to the cytoskeleton. Claudin (Figure 2a), a transmembrane protein of 207–305 amino acids, is the main structural component of the TJs [41]. Different claudin isoforms are expressed in brain endothelial cells, such as claudin-1, -3, -5, and -12; claudin-5 being the most highly expressed in these cells [12]. Occludin (Figure 2a), another tetraspan transmembrane protein of 522 amino acids of TJs, regulates BBB integrity and permeability [42]. The transmembrane TJ proteins are linked to the actin cytoskeleton through the zonula occludens-1 (ZO-1; Figure 2a), a membrane-associated protein [43]. Together, these complex of TJ proteins decrease the paracellular diffusion and limit transcellular activity [12].
The AJs are typically found intermingled with the TJs and contribute to regulating the BBB permeability and leukocyte extravasation [44]. Vascular endothelial (VE)-cadherin is an endothelial-specific integral membrane protein, connected to the cytoskeleton via catenins (Figure 2a). The localization and expression of β-catenin, α-catenin, and p120cas, are essential for the functionality of AJs [40]. Neutrophils and lymphocytes can ingress from the blood into the brain regulated by BBB and low levels of leukocyte adhesion molecules (LAMs) [45]. Therefore, this property may prevent access of immune cells from blood to the brain, affording immunologic benefit in the CNS [45].
BBB integrity is essential for controlling the molecular composition of brain interstitial fluid (ISF), which is indispensable for correct information processing, synaptic operating, functioning, and neuronal connectivity [34]. BBB breakdown could trigger an increase in vascular permeability, reduce CBF and impaired hemodynamic responses [17]. Therefore, BBB dysfunction could facilitate the entry of toxic blood-derived molecules and cells and trigger an immune response associated with neuroinflammation, which ends in degeneration of the NVU (Figure 2b and Figure 3 steps 2c, 3 and 4).

4. Dysfunction of the NVU and BBB in AD Brains

4.1. Bidirectional Pathological Association between Tau and the NVU and BBB

Tau is a microtubule-associated protein that stabilizes microtubules (Figure 2a), polymers of tubulin that form part of the cytoskeleton and provide structure to eukaryotic cells [46]. Tau protein has also been associated with cell signaling, regulation of genomic stability, and synaptic plasticity [47]. Among numerous post-translational modifications, tau is subject to phosphorylation and truncation in AD brain tissue (Figure 2b). Both abnormal changes have been implicated in the accumulation of abnormal tau polymers and paired helical filaments (PHFs) [48,49]. Pathological tau protein leads to microtubule disintegration and NFT formation (Figure 2b) [50,51]. Evidence suggests that dysfunction of the NVU and BBB is triggered by Aβ vascular deposits [52]. However, it has been noted that the presence of pathological tau has been observed as puncta in perivascular spaces in sporadic AD brains [53]. Therefore, dysfunction of the NVU and BBB could trigger tau hyperphosphorylation and, vice versa, tau pathology could induce the NVU/BBB alteration [54] (Figure 2b).
In a transgenic mouse that overexpresses human tau protein carrying the P301L mutation, tau accumulates and eventually leads to BBB disruption [53]. However, the mechanisms by which modified tau could trigger the BBB breakdown are not yet understood. It has been demonstrated that human truncated tau upregulated mRNA expression for several MAPKs (JNK1, p38b, ERK1) and transcription factors (c-Jun, c-Fos, NFkB1, NFkB2). Transcription of pro-inflammatory genes, ultimately leading to the release of proinflammatory cytokines is also increased (Figure 2b). The neuroinflammatory environment has been closely related to BBB dysfunction by increasing permeability, promoting structural changes in brain capillaries, and enhancing the migration of immune cells (Figure 2b) [55]. It has also been suggested that tau-induced activation of glial cells raises the expression of endothelial adhesion molecules and the transport of leukocytes across the BBB, perpetuating the neuroinflammatory environment and exacerbating AD pathology (Figure 2b) [2].
On the other hand, dysfunction of the NVU and BBB could lead to tau pathology. Accumulation of extracellular Aβ induces astrocytes and microglia activation, and the subsequent release of proinflammatory molecules [56]. This neuroinflammatory environment could promote BBB damage and accelerate the development of tau phosphorylation and NFT formation (Figure 2b) [57]. BBB dysfunction can induce pathological tau, and abnormal tau can produce BBB alteration, thus causing a feedback loop (Figure 2b).

4.2. CAA Acts as a Trigger for Dysfunction of the NVU

Dysfunction of the NVU and BBB is closely associated with CAA and may exacerbate AD [58]. CAA is distinguished by the accumulation of Aβ fibrils in the walls of capillaries and small to medium-sized arterial blood vessels caliber of CNS parenchyma and leptomeninges. Besides, CAA is strongly associated with spontaneous intracerebral hemorrhage in older adults and a significant risk factor for age-related cognitive decline [59]. CAA has two presentations: sporadic and hereditary. The sporadic CAA is associated with aging and is a common feature of AD, the amyloid is predominantly composed of the Aβ protein [60]. For hereditary CAA, the nature of the amyloid deposits is associated with the respective underlying mutation in the amyloid precursor protein (APP) gene [60].
Mutations in the APP gene, generally causing single amino acid substitutions, are the triggers of autosomal dominant CAA and early-onset AD. The E693Q (Dutch), L705V (Piedmont), and E693K (Italian) mutations are characterized by severe amyloid angiopathy without NFTs or NPs [61]. Hereditary cerebral hemorrhage with amyloidosis-Dutch type (HCHWA-D), is caused by the E693Q mutation, which causes Aβ aggregation in cerebral and cerebellar meningeal arteries and cerebrocortical arterioles [62]. The histopathological features of HCHWA-D are loss of SMCs, wall thickening, perivascular reactive astrocytes, activated microglia, degenerating neurites, and gemistocytic astrocytes linked to BBB dysfunction [59,63,64,65,66]. These histopathological changes and cognitive deterioration [59] reflect severe vasculopathic changes and severe changes in cellular and structural components of the NVU.

4.3. Dysfunction of Components of the NVU in AD

4.3.1. Perivascular Microglial Activation

Microglia are resident innate immune cells of the CNS [67]. Under physiological conditions, microglia cells are in a “resting or quiescent state” characterized by small-bodied cells with long and thin branches. On the contrary, in pathological conditions, they acquire an “amoeboid or activated state,” they swell and retract their processes and are associated with phagocytic activity and pro-inflammatory cytokine release (Figure 3, step 1) [68]. Furthermore, it has been proposed that microglia are the first cells to degrade both soluble and fibrillar Aβ aggregates (Figure 3, step 1) via receptor interaction (Table 1). This interaction could lead to the activation and production of several toxic molecules [69], and finally, alter the BBB/NVU (Figure 3).

4.3.2. Astrocytic End-Foot Dysfunction

The astrocytic end-foot contributes to arteriolar tone by regulating a constant prostaglandin-E2 (PGE2) flow (Figure 3, step 2a) because of the intracellular Ca2+ fluctuations [95]. Several astrocytic receptors can interact with the Aβ peptide (Table 1), triggering a neuroinflammatory environment and an alteration in the NVU/BBB [96] (Figure 3, step 2a). During the progression of AD, astrocytic end-foot dysfunction could exacerbate the extracellular and vascular accumulation of Aβ by altering its clearance along with the ISF drainage through the glymphatic system [97]. It favors the CSF flow along with the arterial perivascular spaces, and from there to the brain interstitium through the AQP4. Finally, it removes solutes from the neuropil into the cervical and meningeal lymphatic drainage vessels [98]. Dysfunction of the glymphatic system has been observed in AD animal models, most likely because of the dysfunction of AQP4, which increases both Aβ plaque formation and cognitive deficits [24,98]. In humans, genetic variation in AQP4 affects Aβ burden [99], and AD patients with specific AQP4 single nucleotide polymorphism exhibit a rapidly progressive cognitive decline [100]. Histopathological AD brain studies have demonstrated that the loss of perivascular AQP4 localization predicts disease status [101]. Therefore, a dysfunction of the lymphatic system can cause an impairment of Aβ efflux and its accumulation on brain vessels and dysfunction of the NVU (Figure 3, step 2a).

4.3.3. Pericyte Degeneration

Pericytes are essential for regulating the physical and functional BBB properties through proper signaling with brain endothelial cells (BECs). Both platelet-derived growth factor subunit B (PDGF-B) and platelet-derived growth factor receptor-β (PDGFRβ) are closely involved in signaling in these pericytes/BECs [4,102]. Defective signaling leads to BBB disruption, a decrease in CBF, and hypoxia, which in turn triggers neuronal and synaptic dysfunction (Figure 3, step 2b) [102]. These events may further exacerbate both parenchymal and vascular Aβ accumulation [12]. The interaction between Aβ and the pericytes receptors (Table 1) triggers endothelin-1 release from pericytes, resulting in pericyte contraction and subsequently capillary constriction (Figure 3, step 2b) [103]. There is an association between the accumulation of blood-derived proteins (thrombin, fibrinogen, plasminogen, immunoglobulin G, and albumin) and the pericyte degeneration (Figure 3, step 2b) [104]. Thus, pericyte degeneration can cause a decrease in CBF, leading to neurodegeneration.

4.3.4. Endothelial Cell Degeneration and BBB Breakdown

Postmortem studies in AD brains have shown several vascular alterations, including fragmented vessels, string vessels, irregularities in the capillary surface, changes in vessel diameter, thickening, vacuolization, and local rupture of the capillary basement membrane, associated with vascular Aβ accumulation [46]. Also, data from postmortem human brains have demonstrated a substantial reduction in volume associated with CAA [96]. This event might be partly explained because TJs proteins are substrates of vascular-associated matrix metalloproteinase (MMP) activity (Figure 3, step 2c) [105]. Likewise, low levels of mesenchyme homeobox gene 2 (MEOX2) (Figure 3, step 2c) have been reported in AD brains. MEOX2 has been shown to mediate aberrant angiogenic responses, lead to premature capillary pruning [106], and trigger cerebral hypoperfusion and vascular inflammation [107].

4.3.5. Neuronal Cell Death Mechanisms

In AD, cerebral perfusion is impaired because of the compromised ISF drainage due to the destruction of perivascular space and accumulation of Aβ. Aβ can bind to specific neuronal receptors (Table 1) and cause toxicity, induced mainly by oxidative stress. Oligemia can trigger neuronal dysfunction, altering proteins required for synaptic plasticity [108], and favoring anaerobic brain metabolism. Oligemia can also decrease the generation of ATP, necessary for maintaining the Na+, K+-ATPase pump and the required action potentials for normal neuronal excitability [109]. It has been shown that transient depletion of oxygen and glucose loss can cause neuronal excitotoxicity and subsequent neuronal death [58]. Oxygen reductions can alter pH and water-electrolyte balance, leading to edema, white matter lesions, and accumulation of both glutamate and Aβ in the brain [29]. A decrease in the levels of GLUT1 (glucose transporter in neurons) and GLUT3 (glucose transporter in BBB) in AD patients suggests that these may contribute to impaired glucose uptake and metabolism in the brain, which could trigger the phosphorylation of tau protein [46] (Figure 3, step 4).

4.4. The Two-Hit Vascular Hypothesis and Dysfunction of the NVU

According to the two-hit vascular hypothesis of AD, there exists a vascular contribution (hit one) that followed the effect of Aβ accumulation (hit two) [29]. Vascular risk factors, such as diabetes or hypertension, could lead to NVU/BBB dysregulation and, subsequently, CBF reduction (oligemia), initiating a cascade of events that precedes dementia. This defective Aβ clearance has been postulated to trigger increased cerebrovascular Aβ production and aggregation [110]. There are several receptors in the endothelial cells that promote Aβ clearance (Table 2). The primary receptor that regulates Aβ transcytosis is LRP1 (Figure 3, step 2c; Table 2) [111]. In AD, there are significant reductions in LRP1 [112] and SMCs [113] that correlate with an increase in cerebrovascular Aβ [112]. High levels of serum response factor (SRF) and myocardin in SMCs [113] lead to elevated expression of the sterol response element-binding protein 2 (SREBP2), an important transcriptional LRP1 suppressor, triggering LRP1 depletion and reducing Aβ clearance through the BBB (Figure 3, step 2b) [114]. In contrast, RAGE is the receptor for the influx of Aβ through the BBB that transports it from the blood to the brain (Figure 3, step 2c; Table 2) [115], and an increased expression of RAGE in the brain endothelium of AD patients has been reported (Figure 3, step 2c) [104]. Subsequently, oligemia promotes the CBF loss (Figure 3, step3), resulting in endothelial cell death [116]. Finally, the increase in Aβ accumulation (hit two) accelerates neuronal dysfunction, NFT formation, neurodegeneration, and dementia [29] (Figure 3, step 4).

5. Current Status and Challenges for the Pharmacological Treatment of AD

5.1. General Limitations in AD Therapy

One of the main hurdles for developing new drugs for AD is that the BBB limits the access of substances to the brain. The endothelium, one of the main components of the BBB, acts as a physical barrier because of the TJs and AJs. Also, it possesses an enzymatic complex (CYP450, xanthine oxidase, MAO) capable of metabolizing various molecules, reducing their bioactivity in the brain. Likewise, efflux pumps (P-gp, MDR and MRP) placed on the endothelial abluminal surface can release molecules back into the peripheral circulation. In AD, the overexpression of the efflux pump may contribute to the failure of therapeutic trials [150]. It is estimated that around 98% of small and 100% of large molecule drugs, do not cross the BBB and hence do not achieve adequate therapeutic concentrations in the brain [151]. Most drugs are incapable of crossing the BBB due to polarity, size, or lack of endogenous transport pathways [152]. Drug delivery systems (DDS) have emerged as an essential procedure to address these concerns. DDS can preserve molecules from degradation [153] and carry them across membranes by hiding their chemical properties but without modifying them [154]. Nanoparticles, as an example of DDS, serve to release drugs for prolonged periods. Nanoparticle transport by receptor-mediated endocytosis is the main procedure to transport drugs across the BBB [155] and this therapeutic approach will be discussed in a section below.
AD therapeutics has focused on replacing the depletion of acetylcholine levels to improve symptoms in patients. Donepezil, one of the most widely used, is an acetylcholinesterase inhibitor that crosses the BBB via the organic cation transporter [156]. Donepezil has been shown to improve cognitive functions in patients with mild to moderate AD by enhancing the acetylcholine bioavailability [157]. However, a major limitation for donepezil is its P-glycoprotein (P-gp)-mediated efflux [158]. There is a threshold for its use: most patients only tolerate low doses because of its adverse cholinergic side-effects. Besides, patients at a particular phase of the disease do not respond to therapy because there is already neuronal deficit or death and hence absence of synaptic connections [159].

5.2. Therapeutics Associated with the Amyloidogenic Pathway

Aβ aggregation and binding of Aβ to different receptors appears to be a characteristic hallmark of AD. Therapeutic strategies to decrease Aβ production, promote its elimination, and prevent fibrillar aggregation are being developed worldwide. These have targeted the three enzymes associated with the amyloidogenic pathways by increasing the activity of α-secretase or suppressing β- and γ-secretase (Table 2a–c) [160]. Nevertheless, therapies that modulate β- and γ-secretases have been reported to increase the risk of serious adverse events in AD patients [118,161].
There are two types of immunotherapy directed against Aβ peptides: active (Table 2j) and passive immunization (Table 2l) [162]. While both types of immunotherapeutic approaches against Aβ peptides have shown promise in animal models, these are yet to be translated into the clinic [163]. Typically, active immunotherapy consists of an antigen, alone or conjugated to a non-self T helper cell epitope. Active Aβ immunization produces anti-Aβ antibodies that bind to human NPs and can induce long-term antibody [163]. It can also trigger an immune response by activation of Th2 lymphocytes [164]. Passive immunotherapy involves the direct injection of antibodies and does not require the immune system to generate its own antibody response. This therapy has the advantage that it can be directed at a specific epitope and can be stopped if there are any adverse effects [131]. Passive immunization can also promote Aβ clearance by either central or peripheral mechanisms [164]. In the central mechanism, the anti-Aβ antibodies cross the BBB and bind to NPs, triggering an immune response by activating the Th2 lymphocytes. This culminates in the activation of microglial cells responsible for phagocytosis of Aβ and the reduction of amyloid deposits. In the periphery, antibodies can bind and sequester plasma Aβ. When peripheral Aβ is diminished, Aβ efflux from the brain to the bloodstream is favored, reducing the levels of Aβ in the brain [164]. Nevertheless, passive immunization generally requires expensive humanized monoclonal antibodies and repeated injections, making it less suitable for the long-term treatment of AD than active immunization. Repeated injections of antibodies can also lead to the generation of autoantibodies that may neutralize their effect or even trigger side effects such as vasculitis and glomerulonephritis [163].
Circulating antibodies, following immunization, must be able to cross the BBB to exert their cerebral effect. Few studies have analyzed the mechanisms and ability of antibodies directed against Aβ to enter the brain from the bloodstream. These studies have used IgG antibodies, that have demonstrated low permeability and brain accumulation [165,166,167,168]. It has been suggested that these antibodies can cross the BBB through extracellular pathways [169], since most IgG antibodies have an unsaturated blood transport system to the brain, depending on these pathways to enter CNS [169]. Furthermore, it has been proposed that a brain-to-blood efflux system could exist in the BBB for IgG antibodies [170].

5.3. Drugs Targeting Tau Protein

Because pathological tau is both toxic to cells and a mediator of Aβ toxicity [151], targeting tau appears to be a logical therapeutic strategy for AD (Table 2f–i). Several tau aggregation inhibitors have been studied (Table 2f). Methylthioninium, the first tau aggregation inhibitor (TAI) to be discovered [171], was shown to reduce tau pathology and improve cognition in transgenic mouse models of AD and frontotemporal dementia (FTD) [172]. Curcumin, a natural product of the plant Curcuma longa and another TAI, has shown antioxidant and anti-inflammatory properties. However, no significant beneficial effects have been reported in AD subjects receiving curcumin [173].
Tau hyperphosphorylation is observed in AD, and the degree of phosphorylation is reflected by the increased activity of protein kinases [173]. Some kinase inhibitors (Table 2g) prevent cell death and tau phosphorylation, reduce Aβ plaque burden and memory deficits in several models. These compounds can cross the BBB. However, in clinical phases, no clinical improvements have been observed [160,173].
The microtubule stabilizer drugs (Table 2h) have been shown to increase the numbers of microtubules and reduce tau pathology and the number of axons with abnormal morphology in tau transgenic mice. However, some of these drugs have been associated with adverse effects [173].
An antibody targeting the pS396 epitope reduced pathological tau levels and affected the behavioral phenotypes in mice [174,175]. A monoclonal antibody against the epitope in the tau repeat domain inhibited the trans-neuronal propagation of tau aggregation [176]. Several other studies have looked at active and passive immunotherapy approaches in tau mouse models (Table 2i,k) [173]. AADvac1 is an active vaccine consisting of a protein carrier conjugated with a synthetic peptide corresponding to tau [177]. The immunogenicity of AADvac1 in a phase 1 trial has led to a phase 2 trial underway to assess its efficacy in preserving cognitive function [173] (Table 2i). Passive immunization in mouse models has been used to target both extracellular tau and tau aggregates within neurons [173]. Further clinical trials will help establish whether this therapeutic approach is effective in AD (Table 2k).

5.4. Non-Steroidal Anti-Inflammatory Drugs (NSAIDs)

Epidemiologic studies have reported that long-term use of NSAIDs reduced the risk for developing AD [178]. The fenamate NSAIDs inhibit the NLRP3 inflammasome activation and decrease microglial activation [179]. Nevertheless, randomized controlled trials investigating the use of NSAIDs and the risk of developing AD did not reach statistical significance among AD patients and subjects without dementia (Table 2n) [180]. Therefore, the effectiveness of NSAIDs for AD treatment remains uncertain.

6. Therapeutic Strategies Focused on the NVU

Knowledge of the pathophysiology of the NVU in AD is essential for the implementation of new treatments. Emerging therapies are focusing on promoting or maintaining neural stability by either preventing or restoring damage to the NVU.

6.1. Vasculoprotective Effects of Anti-Diabetic, Lipid-Lowering, and Anti-Hypertensive Drugs

According to the two-hit vascular hypothesis of AD, vascular risk factors such as dyslipidemia, obesity, hypertension, and diabetes [181] can trigger dysregulation of the NVU/BBB, leading to dementia [29].
Insulin-resistance has been described in the brain of AD patients [182]. Both insulin and insulin-like growth factor 1 (IGF-1) have multiple brain functions including energy metabolism, trophic support, and synaptic activity [183]. Accumulating evidence suggests that insulin has beneficial effects on the NVU because it is a vasoactive modulator that regulates CBF [184]. Decreased insulin has been associated with endothelial dysfunction, generation of ROS, and excessive free fatty acids released from the adipose tissue [185,186]. Intranasal administration of insulin to the brain has proved feasible in trials with AD patients (Table 3b) [187].
Glucagon-like peptide 1 (GLP-1) analogs are therapeutic agents associated with insulin release and sensitization (Table 3b). The peroxisome proliferator-activated receptors (PPARs) agonists that regulate glucose and lipid metabolism have vasculoprotective and neuroprotective effects, such as regulation of oxidative stress and increasing the CBF and glucose uptake (Table 3b) [204]. Although PPARs were able to improve memory [205], no effect on Aβ pathology was observed in some AD models [204].
Statins (Table 3b) or inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase are a group of medicines used to treat dyslipidemias, that have been shown to have a neuroprotective effect in an animal model [206]. They can restore vascular reactivity and neurovascular coupling, and improve memory, but do not reduce amyloid pathology [207].
Some antihypertensive drugs can improve NVU function (Table 3a) [208]. For example, third-generation β-blockers can improve endothelial function. Calcium channel blockers (CCBs) can have antioxidant and neuroprotective effects, improve endothelial function and vascular inflammation, and attenuate the neuronal deterioration induced by Aβ [208,209]. The angiotensin receptor blocker (ARBs) and angiotensin-converting enzyme (ACE) inhibitors can improve endothelial dysfunction and vascular inflammation [208]. Other antihypertensive drugs include selective angiotensin II type 1 receptor (AT1) blockers. In rodents, AT1 receptor blockers have potential effects on NVU, such as protecting CBF during a stroke and decreasing neuroinflammation and Aβ neurotoxicity [210]. However, they do not affect soluble Aβ or plaque load in the brain [189].

6.2. Drugs for Maintenance of BBB and NVU Integrity

Memantine, an antagonist of extrasynaptic N-methyl-d-aspartate receptors (NMDARs), is currently approved for treatment of AD alone or with acetylcholinesterase inhibitors. NMDARs are ionotropic glutamate receptors that are essential for learning and memory. An increase in NMDAR activation has been related to neuronal dysfunction [211]. Besides, in a mouse model of permanent focal cerebral ischemia, this drug has beneficial effects on NVU because it prevents cerebral ischemia caused by glutamate excitotoxicity, decreases glial activation, and decreases MMP-9 secretion [212]. Therefore, it is considered a drug that in addition to improving cognitive functions can also protect the NVU in AD.
Previous studies have shown that the transforming growth factor-beta (TGF-β), basic fibroblast growth factor (b-FGF), and glial cell-derived neurotrophic factor (GDNF), can promote the BBB integrity (Table 3c) [213].
Proteoglycan NG2, which is a contact adhesion protein that preserves pericyte-brain endothelium interconnections, has been suggested as a possible therapeutic target to maintain correct capillary function [214], and possibly BBB maintenance. Also, drugs that inhibit the metalloproteins effects could keep the BBB integrity [215], limiting the NVU damage.
Notably, a growing body of evidence suggests that interleukin-1β (IL-1β) contributes to BBB dysfunction [216]. Anakinra, an IL-1β receptor antagonist, has been used successfully to treat rheumatoid arthritis [217] and could be a therapeutic target to maintain the BBB integrity in AD (Table 3c). On the other hand, glucocorticoids such as dexamethasone could promote the BBB integrity, triggering the signaling mediated by the glucocorticoid receptor, and regulating the TJs gene expression [218]. Future studies are required to elucidate the beneficial effects of AD.
Multiple sclerosis treatment based on monoclonal antibodies against the α4-integrin receptor on leukocytes [219] (Table 3c) has emerged as a promising therapeutic candidate for avoiding the peripheral immune cell migration into the CNS, reducing neuroinflammation and BBB breakdown in AD.
Targeted inhibition of RAGE–Aβ interaction has been shown to reduce Aβ influx across the BBB, consecutively decreasing the oxidant stress and neuroinflammation environment [82]. However, additional studies with these drugs reporting on long-term outcomes in AD models are required.

6.3. Recent Approaches for Improving the NVU

Molecular Trojan horses (MTJs), nanotechnology, and liposomes have emerged as nano-based DDS. DDS offers several benefits for potential drug molecules. These include the ability to preserve molecules from degradation, transport them through the BBB, and conserve their physicochemical properties, and they are biodegradable, biocompatible, nontoxicity, and non-immunogenic [155]. MTJs are non-invasive approaches for drug delivery of large molecules to the brain, using receptor-mediated transcytosis (RMT) [220].
During neuroinflammation, there is an active change with the regulation of pro-and anti-inflammatory signals [221]. Microglia and astrocytes, which constitute the key elements of the NVU, are activated in several neurological diseases, among them AD. Typically, the pro-inflammatory phenotype (M1/A1) corresponds to dysfunction of the NVU [222,223]. It has been suggested that activated microglia induce reactive astrocytes [224]. Reactive astrocytes (A1) can be prompted by cytokines, such as interleukin-1 alpha (IL-1α), TNF-α, and the complement component subunit 1q (C1q), which are secreted by activated microglia (M1) both in vitro and in vivo [224]. The inhibition of these cytokines might result in repair of the NVU and cognitive improvement in AD patients. Therefore, decoy receptors, such as the human tumor necrosis factor receptor (TNFR), have been proposed as novel therapies for neurological disorders [225]. The human TNFR-II extracellular domain as a fusion protein with a chimeric monoclonal antibody (mAb) against the human insulin receptor (HIR) to permit BBB transport of a TNFR decoy insulin receptor has been considered one of the best candidates for RTM-based therapeutics. Human insulin mAb (HImAb) is around 900% more active and ten times more successful than any human transferrin receptor (TfR) [225]. The HIRmAb can function to transport the TNFR therapeutic decoy receptor across the BBB and provide high-affinity binding to human TNFα, suppressing the cytotoxic effects of this cytokine [225]. These properties may be effective in the restoration of the NVU in AD brains.
Nanotechnology is a new field offering an encouraging perspective in the therapy of various CNS disorders, among them AD, and entails the use of materials that have a dimension of between 1 and 100 nm [155]. Nanoparticles cross the BBB, and therefore allow for certain drugs to be delivered to the brain [226]. Human serum albumin (HSA) nanoparticles seem to be a promising therapeutic approach and would allow the administration of some drugs through the BBB [227]. However, future research is required to assess biosecurity and effectiveness for AD.
There is a diversity of natural compounds unable to cross the BBB [155]. They can be administrated through nanoparticles to be effective in AD therapy due to their neuroprotective activity [155]. These beneficial effects on the NVU could result from the inhibition of the Aβ production (by, for example, regulation of secretase activities),increasing Aβ degradation, inhibition of NFT formation, and reduction of neuroinflammation and oxidative stress [228]. The enhancement of a biodegradable nanoparticle was demonstrated for efficient brain accumulation, protection of astrocytes from oxidative stress and mitochondrial alterations using an animal model [229]. This nanomedicine platform has potential beneficial effects on the NVU such as modulating astrocytes to enhance their neuroprotective activities [229].
Several nanoparticles have shown neuroprotective effects through drugs that modulate inflammation and apoptosis, and the neuronal signaling pathways, by down-regulating pro-inflammatory cytokines [230,231] and caspase-3 activity [232], and up-regulating anti-inflammatory cytokines in animal models [230,231]. Their administration resulted in lower mRNA expression of MMP-9, cyclo-oxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS) [230,231]. All of these molecules are crucial elements for NVU dysfunction [233], and hence promising as targets for AD therapy.
Finally, targeted nanoliposomes and nanoparticles represent a viable and promising strategy for the treatment of AD [234]. They are biocompatible and can carry many therapeutic molecules across the BBB and into NVU cells. Interestingly, immunolabelled liposomes can potentially deliver other novel technologies, such as the mAb MTJs [235]. Modifications of the liposome can include Aβ-targeting ligands (such as curcumin, phosphatidic acid), or retro-inverted peptides, which decrease Aβ and tau aggregation, neuroinflammation, and oxidative stress [234], among others. However, the nonspecific uptake of the cationic liposomes by peripheral tissues and their binding to other proteins limit their therapeutic potential [236]. The intranasal administration of liposomes offers a direct nose-to-brain route. Liposomal encapsulation can facilitate transport across the mucosal barrier. Thus, the intranasal route provides a promising alternative for delivery across the BBB [234].
Additional studies focused on designing new, safe, and effective therapeutic drugs to prevent or restore the NVU damage in AD patients are needed.

7. Conclusions and Perspective

Pathological NVU dysfunction has emerged as an essential element in AD pathogenesis. It has been suggested that vascular amyloid deposits can alter the NVU. The dysfunction of the signaling pathways between each of the cells that make up the NVU is associated with a decrease in CBF, hypoxia, neuroinflammatory status, oxidative stress, and finally, degeneration of these cells. It suggests the need to focus on therapies to prevent or repair damage to the NVU. A significant limitation in the treatment for AD is the BBB presented by the endothelial cells. Many drugs cannot cross BBB or are metabolized, decreasing their bioactivity before reaching the brain. Recent therapeutic approaches, that are still at preclinical phases, such as molecular Trojan horses (MTJs), nanotechnology, and liposomes, have been demonstrated to be of use in getting molecules across the BBB and in restoring dysfunction of the NVU. Because the NVU comprises different cellular and structural components, it becomes a real challenge to target them as a therapeutic target. Nowadays, AD therapy has focused on the interaction of limited parts of the NVU. However, it may only be necessary to impact two of the NVU elements to create a domino effect on other components. Further research is required to better demonstrate the mechanisms involved and determine whether immunotherapies or medications aimed at treating cerebrovascular risk factors for AD are able to reduce brain pathology and improve cognitive function. In this review, we have highlighted the physiological and pathological roles of the NVU to identify where avenues for developing innovative treatments for AD may exist.

Author Contributions

M.P.-H. and J.L.-M., contributed to the conceptualization, writing, review and editing of the manuscript. L.O.S.-R. and C.R.H. contributed to the writing, review and editing of the manuscript. P.A.M.-G., B.B.C.-C., R.A.-P., M.M.V.-R., F.d.l.C., L.G.-R. contributed to the review of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Fondo Nacional de Ciencia y Tecnologia (FONDOCyT) from the Ministry of Higher Education, Science and Technology, Dominican Republic (2015-3A2-127 to M.P.-H.) and (2018-2019-2A3-208 to J.L.-M. and M.P.-H.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors want to express their gratitude to the Mexican families who have donated the brain of their loved ones affected by Alzheimer’s disease and made our research possible. We also want to express our gratitude to the Union Medical University Clinic, Dominican Republic, for their support and collaboration in developing of this research project. This work is dedicated to the memory of José Raúl Mena López †.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

a7nAChRalpha-7 nicotinic acetylcholine receptor
ABCA1ATP binding cassette A1
ABCB1ATP-binding cassette sub-family B member 1
ACEAngiotensin-converting enzyme
ADAlzheimer’s disease
AJAdherent-junction
ApoEApolipoprotein E
APPAmyloid precursor protein
AQP4Aquaporin-4 water channels
ARBAngiotensin receptor blocker
AT1Angiotensin II type 1 receptor
Amyloid β-peptide
BACE1Beta-site APP cleaving enzyme 1
BBBBlood-brain barrier
BDNFBrain-derived neurotrophic factor
BECBrain endothelial cell
b-FGFBasic fibroblast growth factor
C.p.Clinical phase
CxComplement factors
CAACerebral amyloid angiopathy
CBFCerebral blood flow
CCBCalcium channel blocker
CNSCentral nervous system
COX-2Cyclooxygenase-2
CRComplement receptor
CSFCerebrospinal fluid
DDSDrug delivery systems
eNOSEndothelial nitric oxide synthase
fAβFibrillar Aβ
Fc γ RFc γ receptor
FynProto-oncogene tyrosine-protein kinase Fyn
GDNFGlial cell-derived neurotrophic factor
GLP-1Glucagon-like peptide 1
GLUTGlucose transporter
GSK3βGlycogen synthase kinase 3β
HCHWA-DHereditary cerebral hemorrhage with amyloidosis-Dutch type
HImAbHuman insulin mAb
HMG-CoA3-Hydroxy-3-methylglutaryl-CoA
HSAHuman serum albumin
IELInternal elastic lamina
IGF-1Insulin-like growth factor 1
IgGImmunoglobulin G
IL-1αInterleukin-1 alpha
IL-1 βInterleukin-1 beta
IL-1R1Interleukin 1 receptor type 1
iNosinducible nitric oxide synthase
IRInsulin receptor
ISFInterstitial fluid
LAMsLeukocyte adhesion molecules
LRP1Low-density lipoprotein receptor-related protein 1
mAbMonoclonal antibody
mAβMonomeric Aβ
MEOX2Mesenchyme homeobox gene 2
MMPMatrix metalloproteinase
MTJMolecular Trojan horse
NF-kBNuclear factor-κB
NFTNeurofibrillary tangle
NGF Nerve growth factor
NMDARN-Methyl-d-aspartate receptors
NPNeuritic plaque
NSAIDsNon-steroidal anti-inflammatory drugs
NVUNeurovascular unit
oAβOligomeric Aβ
p47PHOXNeutrophil cytosol factor 1
PcpPreclinical phase
PDGF-BPlatelet-derived growth factor subunit B
PDGFRβPlatelet-derived growth factor receptor-β
PGE2Prostaglandin-E2
P-gpP-glycoprotein
PHFPaired helical filament
PI3KPhosphatidylinositol 3-kinase
PVMsPerivascular macrophages
RAGEReceptor for advanced glycation endproducts
RMTReceptor-mediated transcytosis
ROSReactive oxygen species
sAPPαSecreted ectodomain APP alpha
SASSubarachnoid space
SEC-RSerpin-enzyme complex receptor
sLRPSoluble low-density lipoprotein receptor-related protein
SMCSmooth muscle cell
SRAClass A scavenger receptor
SRB2Scavenger receptor class B member 2
SREBP2Sterol response element-binding protein 2
SRFSerum response factor
TfRHuman transferrin receptor
TGF-βTransforming growth factor-beta
TJTight-junction
TLRToll-like receptor
TNF-αTumoral necrosis factor-α
TNFRTumoral necrosis factor receptor
TREM2Triggering receptor expressed on myeloid cells 2
VEGFVascular endothelial growth factor
ZO-1Zonula occludens-1
γ-PPARPeroxisome proliferator-activated receptor-γ

References

  1. Harder, D.R.; Zhang, C.; Gebremedhin, D. Astrocytes function in matching blood flow to metabolic activity. News Physiol. Sci. 2002, 17, 27–31. [Google Scholar] [CrossRef] [PubMed]
  2. Zlokovic, B.V. The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron 2008, 57, 178–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Abbott, N.J.; Friedman, A. Overview and introduction: The blood-brain barrier in health and disease. Epilepsia 2012, 53, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Armulik, A.; Genove, G.; Mae, M.; Nisancioglu, M.H.; Wallgard, E.; Niaudet, C.; He, L.; Norlin, J.; Lindblom, P.; Strittmatter, K.; et al. Pericytes regulate the blood-brain barrier. Nature 2010, 468, 557–561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Winkler, E.A.; Bell, R.D.; Zlokovic, B.V. Central nervous system pericytes in health and disease. Nat. Neurosci. 2011, 14, 1398–1405. [Google Scholar] [CrossRef] [Green Version]
  6. Muoio, V.; Persson, P.B.; Sendeski, M.M. The neurovascular unit—Concept review. Acta Physiol. (Oxford) 2014, 210, 790–798. [Google Scholar] [CrossRef]
  7. Filosa, J.A. Vascular tone and neurovascular coupling: Considerations toward an improved in vitro model. Front. Neuroenerg. 2010, 2, 16. [Google Scholar] [CrossRef] [Green Version]
  8. Fields, R.D.; Stevens-Graham, B. New insights into neuron-glia communication. Science 2002, 298, 556–562. [Google Scholar] [CrossRef] [Green Version]
  9. Yu, X.; Ji, C.; Shao, A. Neurovascular unit dysfunction and neurodegenerative disorders. Front. Neurosci. 2020, 14, 334. [Google Scholar] [CrossRef]
  10. Mucke, L. Neuroscience: Alzheimer’s disease. Nature 2009, 461, 895–897. [Google Scholar] [CrossRef]
  11. Sagare, A.P.; Bell, R.D.; Zlokovic, B.V. Neurovascular defects and faulty amyloid-beta vascular clearance in Alzheimer’s disease. J. Alzheimers Dis. 2013, 33, S87–S100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Yamazaki, Y.; Kanekiyo, T. Blood-brain barrier dysfunction and the pathogenesis of Alzheimer’s disease. Int. J. Mol. Sci. 2017, 18, 1965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Miller, D.L.; Papayannopoulos, I.A.; Styles, J.; Bobin, S.A.; Lin, Y.Y.; Biemann, K.; Iqbal, K. Peptide compositions of the cerebrovascular and senile plaque core amyloid deposits of Alzheimer’s disease. Arch. Biochem. Biophys. 1993, 301, 41–52. [Google Scholar] [CrossRef] [PubMed]
  14. Han, B.H.; Zhou, M.L.; Abousaleh, F.; Brendza, R.P.; Dietrich, H.H.; Koenigsknecht-Talboo, J.; Cirrito, J.R.; Milner, E.; Holtzman, D.M.; Zipfel, G.J. Cerebrovascular dysfunction in amyloid precursor protein transgenic mice: Contribution of soluble and insoluble amyloid-beta peptide, partial restoration via gamma-secretase inhibition. J. Neurosci. 2008, 28, 13542–13550. [Google Scholar] [CrossRef]
  15. Chen, S.J.; Tsai, H.H.; Tsai, L.K.; Tang, S.C.; Lee, B.C.; Liu, H.M.; Yen, R.F.; Jeng, J.S. Advances in cerebral amyloid angiopathy imaging. Ther. Adv. Neurol. Disord. 2019, 12, 1–11. [Google Scholar] [CrossRef]
  16. Xu, F.; Fu, Z.; Dass, S.; Kotarba, A.E.; Davis, J.; Smith, S.O.; van Nostrand, W.E. Cerebral vascular amyloid seeds drive amyloid beta-protein fibril assembly with a distinct anti-parallel structure. Nat. Commun. 2016, 7, 13527. [Google Scholar] [CrossRef] [Green Version]
  17. Kisler, K.; Nelson, A.R.; Montagne, A.; Zlokovic, B.V. Cerebral blood flow regulation and neurovascular dysfunction in Alzheimer disease. Nat. Rev. Neurosci. 2017, 18, 419–434. [Google Scholar] [CrossRef] [Green Version]
  18. Hamel, E. Perivascular nerves and the regulation of cerebrovascular tone. J. Appl. Physiol. 2006, 100, 1059–1064. [Google Scholar] [CrossRef] [Green Version]
  19. Sekiguchi, Y.; Takuwa, H.; Kawaguchi, H.; Kikuchi, T.; Okada, E.; Kanno, I.; Ito, H.; Tomita, Y.; Itoh, Y.; Suzuki, N.; et al. Pial arteries respond earlier than penetrating arterioles to neural activation in the somatosensory cortex in awake mice exposed to chronic hypoxia: An additional mechanism to proximal integration signaling? J. Cereb. Blood Flow Metab. 2014, 34, 1761–1770. [Google Scholar] [CrossRef]
  20. Roggendorf, W.; Cervos-Navarro, J. Ultrastructure of arterioles in the cat brain. Cell Tissue Res. 1977, 178, 495–515. [Google Scholar] [CrossRef]
  21. Zhang, E.T.; Inman, C.B.; Weller, R.O. Interrelationships of the pia mater and the perivascular (Virchow-Robin) spaces in the human cerebrum. J. Anat. 1990, 170, 111–123. [Google Scholar]
  22. Jessen, N.A.; Munk, A.S.; Lundgaard, I.; Nedergaard, M. The glymphatic system: A beginner’s guide. Neurochem. Res. 2015, 40, 2583–2599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Rangroo Thrane, V.; Thrane, A.S.; Plog, B.A.; Thiyagarajan, M.; Iliff, J.J.; Deane, R.; Nagelhus, E.A.; Nedergaard, M. Paravascular microcirculation facilitates rapid lipid transport and astrocyte signaling in the brain. Sci. Rep. 2013, 3, 2582. [Google Scholar] [CrossRef] [PubMed]
  24. Nedergaard, M.; Goldman, S.A. Glymphatic failure as a final common pathway to dementia. Science 2020, 370, 50–56. [Google Scholar] [CrossRef]
  25. Braun, M.; Iliff, J.J. The impact of neurovascular, blood-brain barrier, and glymphatic dysfunction in neurodegenerative and metabolic diseases. Int. Rev. Neurobiol. 2020, 154, 413–436. [Google Scholar] [PubMed]
  26. Hotta, H.; Masamoto, K.; Uchida, S.; Sekiguchi, Y.; Takuwa, H.; Kawaguchi, H.; Shigemoto, K.; Sudo, R.; Tanishita, K.; Ito, H.; et al. Layer-specific dilation of penetrating arteries induced by stimulation of the nucleus basalis of Meynert in the mouse frontal cortex. J. Cereb. Blood Flow Metab. 2013, 33, 1440–1447. [Google Scholar] [CrossRef] [Green Version]
  27. Longden, T.A.; Hill-Eubanks, D.C.; Nelson, M.T. Ion channel networks in the control of cerebral blood flow. J. Cereb. Blood Flow Metab. 2016, 36, 492–512. [Google Scholar] [CrossRef] [Green Version]
  28. Attwell, D.; Buchan, A.M.; Charpak, S.; Lauritzen, M.; Macvicar, B.A.; Newman, E.A. Glial and neuronal control of brain blood flow. Nature 2010, 468, 232–243. [Google Scholar] [CrossRef] [Green Version]
  29. Zlokovic, B.V. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat. Rev. Neurosci. 2011, 12, 723–738. [Google Scholar] [CrossRef]
  30. Damisah, E.C.; Hill, R.A.; Tong, L.; Murray, K.N.; Grutzendler, J. A fluoro-Nissl dye identifies pericytes as distinct vascular mural cells during in vivo brain imaging. Nat. Neurosci. 2017, 20, 1023–1032. [Google Scholar] [CrossRef] [Green Version]
  31. Iadecola, C. The neurovascular unit coming of age: A journey through neurovascular coupling in health and disease. Neuron 2017, 96, 17–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Cuevas, P.; Gutierrez-Diaz, J.A.; Reimers, D.; Dujovny, M.; Diaz, F.G.; Ausman, J.I. Pericyte endothelial gap junctions in human cerebral capillaries. Anat. Embryol. (Berlin) 1984, 170, 155–159. [Google Scholar] [CrossRef] [PubMed]
  33. Zhao, Z.; Nelson, A.R.; Betsholtz, C.; Zlokovic, B.V. Establishment and dysfunction of the blood-brain barrier. Cell 2015, 163, 1064–1078. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Sweeney, M.D.; Sagare, A.P.; Zlokovic, B.V. Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat. Rev. Neurol. 2018, 14, 133–150. [Google Scholar] [CrossRef] [PubMed]
  35. Smith, Q.R. A review of blood-brain barrier transport techniques. Methods Mol. Med. 2003, 89, 193–208. [Google Scholar] [PubMed]
  36. Ohtsuki, S.; Terasaki, T. Contribution of carrier-mediated transport systems to the blood-brain barrier as a supporting and protecting interface for the brain; importance for CNS drug discovery and development. Pharm. Res. 2007, 24, 1745–1758. [Google Scholar] [CrossRef]
  37. Pardridge, W.M. Blood-brain barrier delivery. Drug Discov. Today 2007, 12, 54–61. [Google Scholar] [CrossRef]
  38. Deeken, J.F.; Loscher, W. The blood-brain barrier and cancer: Transporters, treatment, and Trojan horses. Clin. Cancer Res. 2007, 13, 1663–1674. [Google Scholar] [CrossRef] [Green Version]
  39. Hawkins, B.T.; Davis, T.P. The blood-brain barrier/neurovascular unit in health and disease. Pharmacol. Rev. 2005, 57, 173–185. [Google Scholar] [CrossRef]
  40. Bazzoni, G.; Dejana, E. Endothelial cell-to-cell junctions: Molecular organization and role in vascular homeostasis. Physiol. Rev. 2004, 84, 869–901. [Google Scholar] [CrossRef] [Green Version]
  41. Gunzel, D.; Yu, A.S. Claudins and the modulation of tight junction permeability. Physiol. Rev. 2013, 93, 525–569. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Furuse, M.; Hirase, T.; Itoh, M.; Nagafuchi, A.; Yonemura, S.; Tsukita, S.; Tsukita, S. Occludin: A novel integral membrane protein localizing at tight junctions. J. Cell Biol. 1993, 123, 1777–1788. [Google Scholar] [CrossRef] [PubMed]
  43. Itoh, M.; Furuse, M.; Morita, K.; Kubota, K.; Saitou, M.; Tsukita, S. Direct binding of three tight junction-associated MAGUKs, ZO-1, ZO-2, and ZO-3, with the COOH termini of claudins. J. Cell Biol. 1999, 147, 1351–1363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Dejana, E.; Orsenigo, F.; Lampugnani, M.G. The role of adherens junctions and VE-cadherin in the control of vascular permeability. J. Cell Sci. 2008, 121, 2115–2122. [Google Scholar] [CrossRef] [Green Version]
  45. Daneman, R.; Prat, A. The blood-brain barrier. Cold Spring Harb. Perspect. Biol. 2015, 7, a020412. [Google Scholar] [CrossRef] [Green Version]
  46. Michalicova, A.; Banks, W.A.; Legath, J.; Kovac, A. Tauopathies—focus on changes at the neurovascular unit. Curr. Alzheimer Res. 2017, 14, 790–801. [Google Scholar] [CrossRef]
  47. Guo, T.; Noble, W.; Hanger, D.P. Roles of tau protein in health and disease. Acta Neuropathol. 2017, 133, 665–704. [Google Scholar] [CrossRef] [Green Version]
  48. Novak, M.; Kabat, J.; Wischik, C.M. Molecular characterization of the minimal protease resistant tau unit of the Alzheimer’s disease paired helical filament. EMBO J. 1993, 12, 365–370. [Google Scholar] [CrossRef]
  49. Goedert, M. Tau protein and the neurofibrillary pathology of Alzheimer’s disease. Ann. N. Y. Acad. Sci. 1996, 777, 121–131. [Google Scholar] [CrossRef]
  50. Alonso, A.D.; Grundke-Iqbal, I.; Barra, H.S.; Iqbal, K. Abnormal phosphorylation of tau and the mechanism of Alzheimer neurofibrillary degeneration: Sequestration of microtubule-associated proteins 1 and 2 and the disassembly of microtubules by the abnormal tau. Proc. Natl. Acad. Sci. USA 1997, 94, 298–303. [Google Scholar] [CrossRef] [Green Version]
  51. Luna-Munoz, J.; Garcia-Sierra, F.; Falcon, V.; Menendez, I.; Chavez-Macias, L.; Mena, R. Regional conformational change involving phosphorylation of tau protein at the Thr231, precedes the structural change detected by Alz-50 antibody in Alzheimer’s disease. J. Alzheimers Dis. 2005, 8, 29–41. [Google Scholar] [CrossRef] [PubMed]
  52. Greenberg, S.M.; Bacskai, B.J.; Hernandez-Guillamon, M.; Pruzin, J.; Sperling, R.; van Veluw, S.J. Cerebral amyloid angiopathy and Alzheimer disease—One peptide, two pathways. Nat. Rev. Neurol. 2020, 16, 30–42. [Google Scholar] [CrossRef] [PubMed]
  53. Blair, L.J.; Frauen, H.D.; Zhang, B.; Nordhues, B.A.; Bijan, S.; Lin, Y.C.; Zamudio, F.; Hernandez, L.D.; Sabbagh, J.J.; Selenica, M.L.; et al. Tau depletion prevents progressive blood-brain barrier damage in a mouse model of tauopathy. Acta Neuropathol. Commun. 2015, 3, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Ramos-Cejudo, J.; Wisniewski, T.; Marmar, C.; Zetterberg, H.; Blennow, K.; de Leon, M.J.; Fossati, S. Traumatic brain injury and Alzheimer’s disease: The cerebrovascular link. EbioMed. 2018, 28, 21–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Erickson, M.A.; Banks, W.A. Blood-brain barrier dysfunction as a cause and consequence of Alzheimer’s disease. J. Cereb. Blood Flow Metab. 2013, 33, 1500–1513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Heneka, M.T.; Carson, M.J.; El Khoury, J.; Landreth, G.E.; Brosseron, F.; Feinstein, D.L.; Jacobs, A.H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R.M.; et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015, 14, 388–405. [Google Scholar] [CrossRef] [Green Version]
  57. 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, 63, 1223–1234. [Google Scholar] [CrossRef]
  58. Shabir, O.; Berwick, J.; Francis, S.E. Neurovascular dysfunction in vascular dementia, Alzheimer’s and atherosclerosis. BMC Neurosci. 2018, 19, 62. [Google Scholar] [CrossRef] [Green Version]
  59. Gatti, L.; Tinelli, F.; Scelzo, E.; Arioli, F.; Di Fede, G.; Obici, L.; Pantoni, L.; Giaccone, G.; Caroppo, P.; Parati, E.A.; et al. Understanding the Pathophysiology of Cerebral Amyloid Angiopathy. Int. J. Mol. Sci. 2020, 21, 3435. [Google Scholar] [CrossRef]
  60. Attems, J.; Jellinger, K.; Thal, D.R.; van Nostrand, W. Review: Sporadic cerebral amyloid angiopathy. Neuropathol. Appl. Neurobiol. 2011, 37, 75–93. [Google Scholar] [CrossRef]
  61. Bugiani, O.; Giaccone, G.; Rossi, G.; Mangieri, M.; Capobianco, R.; Morbin, M.; Mazzoleni, G.; Cupidi, C.; Marcon, G.; Giovagnoli, A.; et al. Hereditary cerebral hemorrhage with amyloidosis associated with the E693K mutation of APP. Arch. Neurol. 2010, 67, 987–995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Maat-Schieman, M.; Roos, R.; van Duinen, S. Hereditary cerebral hemorrhage with amyloidosis-Dutch type. Neuropathology 2005, 25, 288–297. [Google Scholar] [CrossRef] [PubMed]
  63. Maat-Schieman, M.L.; Yamaguchi, H.; Hegeman-Kleinn, I.M.; Welling-Graafland, C.; Natte, R.; Roos, R.A.; van Duinen, S.G. Glial reactions and the clearance of amyloid beta protein in the brains of patients with hereditary cerebral hemorrhage with amyloidosis-Dutch type. Acta Neuropathol. 2004, 107, 389–398. [Google Scholar] [CrossRef] [PubMed]
  64. Maat-Schieman, M.L.; Yamaguchi, H.; van Duinen, S.G.; Natte, R.; Roos, R.A. Age-related plaque morphology and C-terminal heterogeneity of amyloid beta in Dutch-type hereditary cerebral hemorrhage with amyloidosis. Acta Neuropathol. 2000, 99, 409–419. [Google Scholar] [CrossRef]
  65. Maat-Schieman, M.L.; van Duinen, S.G.; Rozemuller, A.J.; Haan, J.; Roos, R.A. Association of vascular amyloid beta and cells of the mononuclear phagocyte system in hereditary cerebral hemorrhage with amyloidosis (Dutch) and Alzheimer disease. J. Neuropathol. Exp. Neurol. 1997, 56, 273–284. [Google Scholar] [CrossRef] [Green Version]
  66. Tagliavini, F.; Giaccone, G.; Bugiani, O.; Frangione, B. Ubiquitinated neurites are associated with preamyloid and cerebral amyloid beta deposits in patients with hereditary cerebral hemorrhage with amyloidosis Dutch type. Acta Neuropathol. 1993, 85, 267–271. [Google Scholar] [CrossRef]
  67. Ajami, B.; Bennett, J.L.; Krieger, C.; Tetzlaff, W.; Rossi, F.M. Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nat. Neurosci. 2007, 10, 1538–1543. [Google Scholar] [CrossRef]
  68. Arcuri, C.; Mecca, C.; Bianchi, R.; Giambanco, I.; Donato, R. The pathophysiological role of microglia in dynamic surveillance, phagocytosis and structural remodeling of the developing CNS. Front. Mol. Neurosci. 2017, 10, 191. [Google Scholar] [CrossRef] [Green Version]
  69. Moore, K.J.; El Khoury, J.; Medeiros, L.A.; Terada, K.; Geula, C.; Luster, A.D.; Freeman, M.W. A CD36-initiated signaling cascade mediates inflammatory effects of beta-amyloid. J. Biol. Chem. 2002, 277, 47373–47379. [Google Scholar] [CrossRef] [Green Version]
  70. Fonseca, M.I.; Chu, S.; Pierce, A.L.; Brubaker, W.D.; Hauhart, R.E.; Mastroeni, D.; Clarke, E.V.; Rogers, J.; Atkinson, J.P.; Tenner, A.J. Analysis of the putative role of CR1 in Alzheimer’s disease: Genetic association, expression and function. PLoS ONE 2016, 11, e0149792. [Google Scholar] [CrossRef] [Green Version]
  71. Oliveira, L.C.; Kretzschmar, G.C.; Dos Santos, A.C.M.; Camargo, C.M.; Nisihara, R.M.; Farias, T.D.J.; Franke, A.; Wittig, M.; Schmidt, E.; Busch, H.; et al. Complement Receptor 1 (CR1, CD35) Polymorphisms and soluble CR1: A proposed anti-inflammatory role to quench the fire of “Fogo Selvagem” Pemphigus Foliaceus. Front. Immunol. 2019, 10, 2585. [Google Scholar] [CrossRef] [PubMed]
  72. Fu, H.; Liu, B.; Frost, J.L.; Hong, S.; Jin, M.; Ostaszewski, B.; Shankar, G.M.; Costantino, I.M.; Carroll, M.C.; Mayadas, T.N.; et al. Complement component C3 and complement receptor type 3 contribute to the phagocytosis and clearance of fibrillar Abeta by microglia. Glia 2012, 60, 993–1003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Zhang, D.; Hu, X.; Qian, L.; Chen, S.H.; Zhou, H.; Wilson, B.; Miller, D.S.; Hong, J.S. Microglial MAC1 receptor and PI3K are essential in mediating beta-amyloid peptide-induced microglial activation and subsequent neurotoxicity. J. Neuroinflamm. 2011, 8, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Stewart, C.R.; Stuart, L.M.; Wilkinson, K.; van Gils, J.M.; Deng, J.; Halle, A.; Rayner, K.J.; Boyer, L.; Zhong, R.; Frazier, W.A.; et al. CD36 ligands promote sterile inflammation through assembly of a Toll-like receptor 4 and 6 heterodimer. Nat. Immunol. 2010, 11, 155–161. [Google Scholar] [CrossRef] [Green Version]
  75. Reed-Geaghan, E.G.; Savage, J.C.; Hise, A.G.; Landreth, G.E. CD14 and toll-like receptors 2 and 4 are required for fibrillar A{beta}-stimulated microglial activation. J. Neurosci. 2009, 29, 11982–11992. [Google Scholar] [CrossRef] [PubMed]
  76. An, X.Q.; Xi, W.; Gu, C.Y.; Huang, X. Complement protein C5a enhances the beta-amyloid-induced neuro-inflammatory response in microglia in Alzheimer’s disease. Med. Sci. (Paris) 2018, 34, 116–120. [Google Scholar] [CrossRef]
  77. Ager, R.R.; Fonseca, M.I.; Chu, S.H.; Sanderson, S.D.; Taylor, S.M.; Woodruff, T.M.; Tenner, A.J. Microglial C5aR (CD88) expression correlates with amyloid-beta deposition in murine models of Alzheimer’s disease. J. Neurochem. 2010, 113, 389–401. [Google Scholar] [CrossRef] [Green Version]
  78. El Khoury, J.; Hickman, S.E.; Thomas, C.A.; Cao, L.; Silverstein, S.C.; Loike, J.D. Scavenger receptor-mediated adhesion of microglia to beta-amyloid fibrils. Nature 1996, 382, 716–719. [Google Scholar] [CrossRef]
  79. Yang, C.N.; Shiao, Y.J.; Shie, F.S.; Guo, B.S.; Chen, P.H.; Cho, C.Y.; Chen, Y.J.; Huang, F.L.; Tsay, H.J. Mechanism mediating oligomeric Abeta clearance by naive primary microglia. Neurobiol. Dis. 2011, 42, 221–230. [Google Scholar] [CrossRef]
  80. Ganote, C.E.; Sims, M.A. Parallel temperature dependence of contracture-associated enzyme release due to anoxia, 2,4-dinitrophenol (DNP), or caffeine and the calcium paradox. Am. J. Pathol. 1984, 116, 94–106. [Google Scholar]
  81. Marshall, R.J. Bayesian analysis of case-control studies. Stat. Med. 1988, 7, 1223–1230. [Google Scholar] [CrossRef] [PubMed]
  82. Deane, R.; Du Yan, S.; Submamaryan, R.K.; LaRue, B.; Jovanovic, S.; Hogg, E.; Welch, D.; Manness, L.; Lin, C.; Yu, J.; et al. RAGE mediates amyloid-beta peptide transport across the blood-brain barrier and accumulation in brain. Nat. Med. 2003, 9, 907–913. [Google Scholar] [CrossRef] [PubMed]
  83. Ni, R.; Marutle, A.; Nordberg, A. Modulation of alpha7 nicotinic acetylcholine receptor and fibrillar amyloid-beta interactions in Alzheimer’s disease brain. J. Alzheimers Dis. 2013, 33, 841–851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Oz, M.; Lorke, D.E.; Yang, K.H.; Petroianu, G. On the interaction of beta-amyloid peptides and alpha7-nicotinic acetylcholine receptors in Alzheimer’s disease. Curr. Alzheimer Res. 2013, 10, 618–630. [Google Scholar] [CrossRef] [PubMed]
  85. Zhao, W.Q.; De Felice, F.G.; Fernandez, S.; Chen, H.; Lambert, M.P.; Quon, M.J.; Krafft, G.A.; Klein, W.L. Amyloid beta oligomers induce impairment of neuronal insulin receptors. FASEB J. 2008, 22, 246–260. [Google Scholar] [CrossRef] [Green Version]
  86. Xie, L.; Helmerhorst, E.; Taddei, K.; Plewright, B.; Van Bronswijk, W.; Martins, R. Alzheimer’s beta-amyloid peptides compete for insulin binding to the insulin receptor. J. Neurosci. 2002, 22, RC221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Boland, K.; Behrens, M.; Choi, D.; Manias, K.; Perlmutter, D.H. The serpin-enzyme complex receptor recognizes soluble, nontoxic amyloid-beta peptide but not aggregated, cytotoxic amyloid-beta peptide. J. Biol. Chem. 1996, 271, 18032–18044. [Google Scholar] [CrossRef] [Green Version]
  88. Lai, A.Y.; McLaurin, J. Mechanisms of amyloid-beta peptide uptake by neurons: The role of lipid rafts and lipid raft-associated proteins. Int. J. Alzheimers Dis. 2010, 2011, 548380. [Google Scholar] [CrossRef] [Green Version]
  89. Zhao, Y.; Wu, X.; Li, X.; Jiang, L.L.; Gui, X.; Liu, Y.; Sun, Y.; Zhu, B.; Pina-Crespo, J.C.; Zhang, M.; et al. TREM2 is a receptor for beta-amyloid that mediates microglial function. Neuron 2018, 97, 1023–1031.e7. [Google Scholar] [CrossRef] [Green Version]
  90. Hsieh, C.L.; Koike, M.; Spusta, S.C.; Niemi, E.C.; Yenari, M.; Nakamura, M.C.; Seaman, W.E. A role for TREM2 ligands in the phagocytosis of apoptotic neuronal cells by microglia. J. Neurochem. 2009, 109, 1144–1156. [Google Scholar] [CrossRef] [Green Version]
  91. Ma, Q.; Zhao, Z.; Sagare, A.P.; Wu, Y.; Wang, M.; Owens, N.C.; Verghese, P.B.; Herz, J.; Holtzman, D.M.; Zlokovic, B.V. Blood-brain barrier-associated pericytes internalize and clear aggregated amyloid-beta42 by LRP1-dependent apolipoprotein E isoform-specific mechanism. Mol. Neurodegener. 2018, 13, 57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Wilhelmus, M.M.; Otte-Holler, I.; van Triel, J.J.; Veerhuis, R.; Maat-Schieman, M.L.; Bu, G.; de Waal, R.M.; Verbeek, M.M. Lipoprotein receptor-related protein-1 mediates amyloid-beta-mediated cell death of cerebrovascular cells. Am. J. Pathol. 2007, 171, 1989–1999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Saint-Pol, J.; Vandenhaute, E.; Boucau, M.C.; Candela, P.; Dehouck, L.; Cecchelli, R.; Dehouck, M.P.; Fenart, L.; Gosselet, F. Brain pericytes ABCA1 expression mediates cholesterol efflux but not cellular amyloid-beta peptide accumulation. J. Alzheimers Dis. 2012, 30, 489–503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Kuhnke, D.; Jedlitschky, G.; Grube, M.; Krohn, M.; Jucker, M.; Mosyagin, I.; Cascorbi, I.; Walker, L.C.; Kroemer, H.K.; Warzok, R.W.; et al. MDR1-P-Glycoprotein (ABCB1) Mediates Transport of Alzheimer’s amyloid-beta peptides-implications for the mechanisms of Abeta clearance at the blood-brain barrier. Brain Pathol. 2007, 17, 347–353. [Google Scholar] [CrossRef] [PubMed]
  95. Rosenegger, D.G.; Tran, C.H.; Wamsteeker Cusulin, J.I.; Gordon, G.R. Tonic local brain blood flow control by astrocytes independent of phasic neurovascular coupling. J. Neurosci. 2015, 35, 13463–13474. [Google Scholar] [CrossRef] [PubMed]
  96. Carrano, A.; Hoozemans, J.J.; van der Vies, S.M.; van Horssen, J.; de Vries, H.E.; Rozemuller, A.J. Neuroinflammation and blood-brain barrier changes in capillary amyloid angiopathy. Neurodegener. Dis. 2012, 10, 329–331. [Google Scholar] [CrossRef]
  97. Bakker, E.N.; Bacskai, B.J.; Arbel-Ornath, M.; Aldea, R.; Bedussi, B.; Morris, A.W.; Weller, R.O.; Carare, R.O. Lymphatic clearance of the brain: Perivascular, paravascular and significance for neurodegenerative diseases. Cell Mol. Neurobiol. 2016, 36, 181–194. [Google Scholar] [CrossRef] [Green Version]
  98. Rasmussen, M.K.; Mestre, H.; Nedergaard, M. The glymphatic pathway in neurological disorders. Lancet Neurol. 2018, 17, 1016–1024. [Google Scholar] [CrossRef] [Green Version]
  99. Ulv Larsen, S.M.; Landolt, H.P.; Berger, W.; Nedergaard, M.; Knudsen, G.M.; Holst, S.C. Haplotype of the astrocytic water channel AQP4 is associated with slow wave energy regulation in human NREM sleep. PLoS Biol. 2020, 18, e3000623. [Google Scholar] [CrossRef]
  100. Burfeind, K.G.; Murchison, C.F.; Westaway, S.K.; Simon, M.J.; Erten-Lyons, D.; Kaye, J.A.; Quinn, J.F.; Iliff, J.J. The effects of noncoding aquaporin-4 single-nucleotide polymorphisms on cognition and functional progression of Alzheimer’s disease. Alzheimers Dement. (NY) 2017, 3, 348–359. [Google Scholar] [CrossRef]
  101. Zeppenfeld, D.M.; Simon, M.; Haswell, J.D.; D’Abreo, D.; Murchison, C.; Quinn, J.F.; Grafe, M.R.; Woltjer, R.L.; Kaye, J.; Iliff, J.J. Association of perivascular localization of aquaporin-4 with cognition and Alzheimer disease in aging brains. JAMA Neurol. 2017, 74, 91–99. [Google Scholar] [CrossRef] [PubMed]
  102. Bell, R.D.; Winkler, E.A.; Sagare, A.P.; Singh, I.; LaRue, B.; Deane, R.; Zlokovic, B.V. Pericytes control key neurovascular functions and neuronal phenotype in the adult brain and during brain aging. Neuron 2010, 68, 409–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Alcendor, D.J. Interactions between smyloid-beta proteins and human brain pericytes: Implications for the pathobiology of Alzheimer’s disease. J. Clin. Med. 2020, 9, 1490. [Google Scholar] [CrossRef] [PubMed]
  104. Nelson, A.R.; Sweeney, M.D.; Sagare, A.P.; Zlokovic, B.V. Neurovascular dysfunction and neurodegeneration in dementia and Alzheimer’s disease. Biochim. Biophys. Acta 2016, 1862, 887–900. [Google Scholar] [CrossRef] [PubMed]
  105. Rosenberg, G.A. Matrix metalloproteinases and their multiple roles in neurodegenerative diseases. Lancet Neurol. 2009, 8, 205–216. [Google Scholar] [CrossRef]
  106. Wu, Z.; Guo, H.; Chow, N.; Sallstrom, J.; Bell, R.D.; Deane, R.; Brooks, A.I.; Kanagala, S.; Rubio, A.; Sagare, A.; et al. Role of the MEOX2 homeobox gene in neurovascular dysfunction in Alzheimer disease. Nat. Med. 2005, 11, 959–965. [Google Scholar] [CrossRef] [PubMed]
  107. Ogunshola, O.O.; Antoniou, X. Contribution of hypoxia to Alzheimer’s disease: Is HIF-1alpha a mediator of neurodegeneration? Cell Mol. Life Sci. 2009, 66, 3555–3563. [Google Scholar] [CrossRef] [Green Version]
  108. Iadecola, C. Neurovascular regulation in the normal brain and in Alzheimer’s disease. Nat. Rev. Neurosci. 2004, 5, 347–360. [Google Scholar] [CrossRef]
  109. Iadecola, C. The pathobiology of vascular dementia. Neuron 2013, 80, 844–866. [Google Scholar] [CrossRef] [Green Version]
  110. Mawuenyega, K.G.; Sigurdson, W.; Ovod, V.; Munsell, L.; Kasten, T.; Morris, J.C.; Yarasheski, K.E.; Bateman, R.J. Decreased clearance of CNS beta-amyloid in Alzheimer’s disease. Science 2010, 330, 1774. [Google Scholar] [CrossRef] [Green Version]
  111. Tarasoff-Conway, J.M.; Carare, R.O.; Osorio, R.S.; Glodzik, L.; Butler, T.; Fieremans, E.; Axel, L.; Rusinek, H.; Nicholson, C.; Zlokovic, B.V.; et al. Clearance systems in the brain-implications for Alzheimer disease. Nat. Rev. Neurol. 2015, 11, 457–470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Donahue, J.E.; Flaherty, S.L.; Johanson, C.E.; Duncan, J.A., 3rd; Silverberg, G.D.; Miller, M.C.; Tavares, R.; Yang, W.; Wu, Q.; Sabo, E.; et al. RAGE, LRP-1, and amyloid-beta protein in Alzheimer’s disease. Acta Neuropathol. 2006, 112, 405–415. [Google Scholar] [CrossRef] [PubMed]
  113. Bell, R.D.; Deane, R.; Chow, N.; Long, X.; Sagare, A.; Singh, I.; Streb, J.W.; Guo, H.; Rubio, A.; Van Nostrand, W.; et al. SRF and myocardin regulate LRP-mediated amyloid-beta clearance in brain vascular cells. Nat. Cell Biol. 2009, 11, 143–153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Sagare, A.P.; Bell, R.D.; Zlokovic, B.V. Neurovascular dysfunction and faulty amyloid beta-peptide clearance in Alzheimer disease. Cold Spring Harb. Perspect. Med. 2012, 2, a011452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Deane, R.; Singh, I.; Sagare, A.P.; Bell, R.D.; Ross, N.T.; LaRue, B.; Love, R.; Perry, S.; Paquette, N.; Deane, R.J.; et al. A multimodal RAGE-specific inhibitor reduces amyloid beta-mediated brain disorder in a mouse model of Alzheimer disease. J. Clin. Investig. 2012, 122, 1377–1392. [Google Scholar] [CrossRef] [Green Version]
  116. Hunter, J.M.; Kwan, J.; Malek-Ahmadi, M.; Maarouf, C.L.; Kokjohn, T.A.; Belden, C.; Sabbagh, M.N.; Beach, T.G.; Roher, A.E. Morphological and pathological evolution of the brain microcirculation in aging and Alzheimer’s disease. PLoS ONE 2012, 7, e36893. [Google Scholar] [CrossRef]
  117. Li, J.M.; Huang, L.L.; Liu, F.; Tang, B.S.; Yan, X.X. Can brain impermeable BACE1 inhibitors serve as anti-CAA medicine? BMC Neurol. 2017, 17, 163. [Google Scholar] [CrossRef] [Green Version]
  118. Vassar, R. BACE1 inhibitor drugs in clinical trials for Alzheimer’s disease. Alzheimers. Res. Ther. 2014, 6, 89. [Google Scholar] [CrossRef] [Green Version]
  119. Chris Min, K.; Dockendorf, M.F.; Palcza, J.; Tseng, J.; Ma, L.; Stone, J.A.; Kleijn, H.J.; Hodsman, P.; Masuo, K.; Tanen, M.; et al. Pharmacokinetics and Pharmacodynamics of the BACE1 Inhibitor verubecestat (MK-8931) in healthy Japanese adults: A randomized, placebo-controlled study. Clin. Pharmacol. Ther. 2019, 105, 1234–1243. [Google Scholar] [CrossRef]
  120. Vandenberghe, R.; Riviere, M.E.; Caputo, A.; Sovago, J.; Maguire, R.P.; Farlow, M.; Marotta, G.; Sanchez-Valle, R.; Scheltens, P.; Ryan, J.M.; et al. Active Abeta immunotherapy CAD106 in Alzheimer’s disease: A phase 2b study. Alzheimers Dement. (NY) 2017, 3, 10–22. [Google Scholar] [CrossRef]
  121. Folch, J.; Ettcheto, M.; Petrov, D.; Abad, S.; Pedros, I.; Marin, M.; Olloquequi, J.; Camins, A. Review of the advances in treatment for Alzheimer disease: Strategies for combating beta-amyloid protein. Neurologia 2018, 33, 47–58. [Google Scholar] [CrossRef] [PubMed]
  122. Veeraraghavalu, K.; Zhang, C.; Zhang, X.; Tanzi, R.E.; Sisodia, S.S. Age-dependent, non-cell-autonomous deposition of amyloid from synthesis of beta-amyloid by cells other than excitatory neurons. J. Neurosci. 2014, 34, 3668–3673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Henley, D.B.; May, P.C.; Dean, R.A.; Siemers, E.R. Development of semagacestat (LY450139), a functional gamma-secretase inhibitor, for the treatment of Alzheimer’s disease. Expert Opin. Pharmacother. 2009, 10, 1657–1664. [Google Scholar] [CrossRef] [PubMed]
  124. Coric, V.; Salloway, S.; van Dyck, C.H.; Dubois, B.; Andreasen, N.; Brody, M.; Curtis, C.; Soininen, H.; Thein, S.; Shiovitz, T.; et al. Targeting prodromal Alzheimer disease with avagacestat: A randomized clinical trial. JAMA Neurol. 2015, 72, 1324–1333. [Google Scholar] [CrossRef] [PubMed]
  125. Wu, Y.; Cui, J. (-)-Epigallocatechin-3-gallate provides neuroprotection via AMPK activation against traumatic brain injury in a mouse model. Naunyn Schmiedebergs Arch. Pharmacol. 2020, 393, 2209–2220. [Google Scholar] [CrossRef] [PubMed]
  126. Mori, T.; Koyama, N.; Tan, J.; Segawa, T.; Maeda, M.; Town, T. Combined treatment with the phenolics (-)-epigallocatechin-3-gallate and ferulic acid improves cognition and reduces Alzheimer-like pathology in mice. J. Biol. Chem. 2019, 294, 2714–2731. [Google Scholar] [CrossRef] [Green Version]
  127. Raghuvanshi, R.; Bharate, S.B. Preclinical and clinical studies on bryostatins, a class of marine-derived protein kinase C modulators: A mini-review. Curr. Top Med. Chem. 2020, 20, 1124–1135. [Google Scholar] [CrossRef]
  128. Yiannopoulou, K.G.; Papageorgiou, S.G. Current and future treatments in Alzheimer disease: An update. J. Cent. Nerv. Syst. Dis. 2020, 12. [Google Scholar] [CrossRef] [Green Version]
  129. Talaga, P. Beta-amyloid aggregation inhibitors for the treatment of Alzheimer’s disease: Dream or reality? Mini Rev. Med. Chem. 2001, 1, 175–186. [Google Scholar] [CrossRef]
  130. Santa-Maria, I.; Hernandez, F.; Del Rio, J.; Moreno, F.J.; Avila, J. Tramiprosate, a drug of potential interest for the treatment of Alzheimer’s disease, promotes an abnormal aggregation of tau. Mol. Neurodegener. 2007, 2, 17. [Google Scholar] [CrossRef] [Green Version]
  131. Sabbagh, M.N.; Agro, A.; Bell, J.; Aisen, P.S.; Schweizer, E.; Galasko, D. PF-04494700, an oral inhibitor of receptor for advanced glycation end products (RAGE), in Alzheimer disease. Alzheimer Dis. Assoc. Disord. 2011, 25, 206–212. [Google Scholar] [CrossRef] [PubMed]
  132. Folch, J.; Petrov, D.; Ettcheto, M.; Abad, S.; Sanchez-Lopez, E.; Garcia, M.L.; Olloquequi, J.; Beas-Zarate, C.; Auladell, C.; Camins, A. Current research therapeutic strategies for Alzheimer’s disease treatment. Neural. Plast. 2016, 2016, 8501693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Medina, M. An overview on the clinical development of tau-based therapeutics. Int. J. Mol. Sci. 2018, 19, 1160. [Google Scholar] [CrossRef] [Green Version]
  134. Turner, R.S.; Hebron, M.L.; Lawler, A.; Mundel, E.E.; Yusuf, N.; Starr, J.N.; Anjum, M.; Pagan, F.; Torres-Yaghi, Y.; Shi, W.; et al. Nilotinib effects on safety, tolerability, and biomarkers in Alzheimer’s disease. Ann. Neurol. 2020, 88, 183–194. [Google Scholar] [CrossRef] [PubMed]
  135. Pasquier, F.; Sadowsky, C.; Holstein, A.; Leterme Gle, P.; Peng, Y.; Jackson, N.; Fox, N.C.; Ketter, N.; Liu, E.; Ryan, J.M.; et al. Two phase 2 multiple ascending-dose studies of vanutide cridificar (ACC-001) and QS-21 adjuvant in mild-to-moderate Alzheimer’s disease. J. Alzheimers Dis. 2016, 51, 1131–1143. [Google Scholar] [CrossRef] [PubMed]
  136. Harigaya, Y.; Saido, T.C.; Eckman, C.B.; Prada, C.M.; Shoji, M.; Younkin, S.G. Amyloid beta protein starting pyroglutamate at position 3 is a major component of the amyloid deposits in the Alzheimer’s disease brain. Biochem. Biophys. Res. Commun. 2000, 276, 422–427. [Google Scholar] [CrossRef] [PubMed]
  137. Pike, C.J.; Overman, M.J.; Cotman, C.W. Amino-terminal deletions enhance aggregation of beta-amyloid peptides in vitro. J. Biol. Chem. 1995, 270, 23895–23898. [Google Scholar] [CrossRef] [Green Version]
  138. Wilcock, D.M.; Colton, C.A. Immunotherapy, vascular pathology, and microhemorrhages in transgenic mice. CNS Neurol. Disord. Drug Targets 2009, 8, 50–64. [Google Scholar] [CrossRef] [Green Version]
  139. Budd Haeberlein, S.; O’Gorman, J.; Chiao, P.; Bussiere, T.; von Rosenstiel, P.; Tian, Y.; Zhu, Y.; von Hehn, C.; Gheuens, S.; Skordos, L.; et al. Clinical development of aducanumab, an anti-Abeta human monoclonal antibody being investigated for the treatment of early Alzheimer’s disease. J. Prev. Alzheimers Dis. 2017, 4, 255–263. [Google Scholar]
  140. Crehan, H.; Liu, B.; Kleinschmidt, M.; Rahfeld, J.U.; Le, K.X.; Caldarone, B.J.; Frost, J.L.; Hettmann, T.; Hutter-Paier, B.; O’Nuallain, B.; et al. Effector function of anti-pyroglutamate-3 Abeta antibodies affects cognitive benefit, glial activation and amyloid clearance in Alzheimer’s-like mice. Alzheimers. Res. Ther. 2020, 12, 12. [Google Scholar] [CrossRef]
  141. Samadi, H.; Sultzer, D. Solanezumab for Alzheimer’s disease. Expert Opin. Biol. Ther. 2011, 11, 787–798. [Google Scholar] [CrossRef] [PubMed]
  142. Yang, T.; Dang, Y.; Ostaszewski, B.; Mengel, D.; Steffen, V.; Rabe, C.; Bittner, T.; Walsh, D.M.; Selkoe, D.J. Target engagement in an alzheimer trial: Crenezumab lowers amyloid beta oligomers in cerebrospinal fluid. Ann. Neurol. 2019, 86, 215–224. [Google Scholar] [CrossRef] [PubMed]
  143. Pradier, L.; Blanchard-Bregeon, V.; Bohme, A.; Debeir, T.; Menager, J.; Benoit, P.; Barneoud, P.; Taupin, V.; Bertrand, P.; Dugay, P.; et al. SAR228810: An antibody for protofibrillar amyloid beta peptide designed to reduce the risk of amyloid-related imaging abnormalities (ARIA). Alzheimers. Res. Ther. 2018, 10, 117. [Google Scholar] [CrossRef] [PubMed]
  144. Andreasen, N.; Simeoni, M.; Ostlund, H.; Lisjo, P.I.; Fladby, T.; Loercher, A.E.; Byrne, G.J.; Murray, F.; Scott-Stevens, P.T.; Wallin, A.; et al. First administration of the Fc-attenuated anti-beta amyloid antibody GSK933776 to patients with mild Alzheimer’s disease: A randomized, placebo-controlled study. PLoS ONE 2015, 10, e0098153. [Google Scholar] [CrossRef] [PubMed]
  145. Landen, J.W.; Andreasen, N.; Cronenberger, C.L.; Schwartz, P.F.; Borjesson-Hanson, A.; Ostlund, H.; Sattler, C.A.; Binneman, B.; Bednar, M.M. Ponezumab in mild-to-moderate Alzheimer’s disease: Randomized phase II PET-PIB study. Alzheimers Dement. (NY) 2017, 3, 393–401. [Google Scholar] [CrossRef]
  146. Sollvander, S.; Nikitidou, E.; Gallasch, L.; Zysk, M.; Soderberg, L.; Sehlin, D.; Lannfelt, L.; Erlandsson, A. The Abeta protofibril selective antibody mAb158 prevents accumulation of Abeta in astrocytes and rescues neurons from Abeta-induced cell death. J. Neuroinflamm. 2018, 15, 98. [Google Scholar] [CrossRef]
  147. Loeffler, D.A. Intravenous immunoglobulin and Alzheimer’s disease: What now? J. Neuroinflamm. 2013, 10, 70. [Google Scholar] [CrossRef] [Green Version]
  148. Sano, M. Tarenflurbil: Mechanisms and myths. Arch. Neurol. 2010, 67, 750–752. [Google Scholar] [CrossRef]
  149. Sivilia, S.; Lorenzini, L.; Giuliani, A.; Gusciglio, M.; Fernandez, M.; Baldassarro, V.A.; Mangano, C.; Ferraro, L.; Pietrini, V.; Baroc, M.F.; et al. Multi-target action of the novel anti-Alzheimer compound CHF5074: In vivo study of long term treatment in Tg2576 mice. BMC Neurosci. 2013, 14, 44. [Google Scholar] [CrossRef] [Green Version]
  150. Wijesuriya, H.C.; Bullock, J.Y.; Faull, R.L.; Hladky, S.B.; Barrand, M.A. ABC efflux transporters in brain vasculature of Alzheimer’s subjects. Brain Res. 2010, 1358, 228–238. [Google Scholar] [CrossRef]
  151. Ghose, A.K.; Viswanadhan, V.N.; Wendoloski, J.J. A knowledge-based approach in designing combinatorial or medicinal chemistry libraries for drug discovery. 1. A qualitative and quantitative characterization of known drug databases. J. Comb. Chem. 1999, 1, 55–68. [Google Scholar] [CrossRef] [PubMed]
  152. Pardridge, W.M. Molecular Trojan horses for blood-brain barrier drug delivery. Discov. Med. 2006, 6, 139–143. [Google Scholar] [CrossRef] [PubMed]
  153. Park, K. Drug delivery of the future: Chasing the invisible gorilla. J. Control. Release 2016, 240, 2–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Saraiva, C.; Praca, C.; Ferreira, R.; Santos, T.; Ferreira, L.; Bernardino, L. Nanoparticle-mediated brain drug delivery: Overcoming blood-brain barrier to treat neurodegenerative diseases. J. Control. Release 2016, 235, 34–47. [Google Scholar] [CrossRef] [Green Version]
  155. Ramalho, M.J.; Andrade, S.; Loureiro, J.A.; do Carmo Pereira, M. Nanotechnology to improve the Alzheimer’s disease therapy with natural compounds. Drug Deliv. Transl. Res. 2020, 10, 380–402. [Google Scholar] [CrossRef] [PubMed]
  156. Kim, M.H.; Maeng, H.J.; Yu, K.H.; Lee, K.R.; Tsuruo, T.; Kim, D.D.; Shim, C.K.; Chung, S.J. Evidence of carrier-mediated transport in the penetration of donepezil into the rat brain. J. Pharm. Sci. 2010, 99, 1548–1566. [Google Scholar] [CrossRef]
  157. Brousseau, G.; Rourke, B.P.; Burke, B. Acetylcholinesterase inhibitors, neuropsychiatric symptoms, and Alzheimer’s disease subtypes: An alternate hypothesis to global cognitive enhancement. Exp. Clin. Psychopharmacol. 2007, 15, 546–554. [Google Scholar] [CrossRef]
  158. Ishiwata, K.; Kawamura, K.; Yanai, K.; Hendrikse, N.H. In vivo evaluation of P-glycoprotein modulation of 8 PET radioligands used clinically. J. Nucl. Med. 2007, 48, 81–87. [Google Scholar]
  159. Scheff, S.W.; Price, D.A.; Schmitt, F.A.; Mufson, E.J. Hippocampal synaptic loss in early Alzheimer’s disease and mild cognitive impairment. Neurobiol. Aging 2006, 27, 1372–1384. [Google Scholar] [CrossRef]
  160. Dong, Y.; Li, X.; Cheng, J.; Hou, L. Drug development for Alzheimer’s disease: Microglia induced neuroinflammation as a target? Int. J. Mol. Sci. 2019, 20, 558. [Google Scholar] [CrossRef] [Green Version]
  161. Golde, T.E.; Koo, E.H.; Felsenstein, K.M.; Osborne, B.A.; Miele, L. gamma-Secretase inhibitors and modulators. Biochim. Biophys. Acta 2013, 1828, 2898–2907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Gilman, S.; Koller, M.; Black, R.S.; Jenkins, L.; Griffith, S.G.; Fox, N.C.; Eisner, L.; Kirby, L.; Rovira, M.B.; Forette, F.; et al. Clinical effects of Abeta immunization (AN1792) in patients with AD in an interrupted trial. Neurology 2005, 64, 1553–1562. [Google Scholar] [CrossRef] [PubMed]
  163. Lemere, C.A. Immunotherapy for Alzheimer’s disease: Hoops and hurdles. Mol. Neurodegener. 2013, 8, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. DeMattos, R.B.; Bales, K.R.; Cummins, D.J.; Dodart, J.C.; Paul, S.M.; Holtzman, D.M. Peripheral anti-A beta antibody alters CNS and plasma A beta clearance and decreases brain A beta burden in a mouse model of Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2001, 98, 8850–8855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Banks, W.A.; Pagliari, P.; Nakaoke, R.; Morley, J.E. Effects of a behaviorally active antibody on the brain uptake and clearance of amyloid beta proteins. Peptides 2005, 26, 287–294. [Google Scholar] [CrossRef] [PubMed]
  166. Banks, W.A.; Terrell, B.; Farr, S.A.; Robinson, S.M.; Nonaka, N.; Morley, J.E. Passage of amyloid beta protein antibody across the blood-brain barrier in a mouse model of Alzheimer’s disease. Peptides 2002, 23, 2223–2226. [Google Scholar] [CrossRef]
  167. Poduslo, J.F.; Curran, G.L.; Berg, C.T. Macromolecular permeability across the blood-nerve and blood-brain barriers. Proc. Natl. Acad. Sci. USA 1994, 91, 5705–5709. [Google Scholar] [CrossRef] [Green Version]
  168. Kozlowski, G.P.; Sterzl, I.; Nilaver, G. Localization patterns for immunoglobulins and albumins in the brain suggest diverse mechanisms for their transport across the blood-brain barrier (BBB). Prog. Brain Res. 1992, 91, 149–154. [Google Scholar]
  169. Banks, W.A. Are the extracellular pathways a conduit for the delivery of therapeutics to the brain? Curr. Pharm. Des. 2004, 10, 1365–1370. [Google Scholar] [CrossRef]
  170. Schlachetzki, F.; Zhu, C.; Pardridge, W.M. Expression of the neonatal Fc receptor (FcRn) at the blood-brain barrier. J. Neurochem. 2002, 81, 203–206. [Google Scholar] [CrossRef]
  171. Wischik, C.M.; Edwards, P.C.; Lai, R.Y.; Roth, M.; Harrington, C.R. Selective inhibition of Alzheimer disease-like tau aggregation by phenothiazines. Proc. Natl. Acad. Sci. USA 1996, 93, 11213–11218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Melis, V.; Magbagbeolu, M.; Rickard, J.E.; Horsley, D.; Davidson, K.; Harrington, K.A.; Goatman, K.; Goatman, E.A.; Deiana, S.; Close, S.P.; et al. Effects of oxidized and reduced forms of methylthioninium in two transgenic mouse tauopathy models. Behav. Pharmacol. 2015, 26, 353–368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Congdon, E.E.; Sigurdsson, E.M. Tau-targeting therapies for Alzheimer disease. Nat. Rev. Neurol. 2018, 14, 399–415. [Google Scholar] [CrossRef] [PubMed]
  174. Asuni, A.A.; Boutajangout, A.; Quartermain, D.; Sigurdsson, E.M. Immunotherapy targeting pathological tau conformers in a tangle mouse model reduces brain pathology with associated functional improvements. J. Neurosci. 2007, 27, 9115–9129. [Google Scholar] [CrossRef] [PubMed]
  175. Boutajangout, A.; Ingadottir, J.; Davies, P.; Sigurdsson, E.M. Passive immunization targeting pathological phospho-tau protein in a mouse model reduces functional decline and clears tau aggregates from the brain. J. Neurochem. 2011, 118, 658–667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Kfoury, N.; Holmes, B.B.; Jiang, H.; Holtzman, D.M.; Diamond, M.I. Trans-cellular propagation of Tau aggregation by fibrillar species. J. Biol. Chem. 2012, 287, 19440–19451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Kontsekova, E.; Zilka, N.; Kovacech, B.; Novak, P.; Novak, M. First-in-man tau vaccine targeting structural determinants essential for pathological tau-tau interaction reduces tau oligomerisation and neurofibrillary degeneration in an Alzheimer’s disease model. Alzheimers. Res. Ther. 2014, 6, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Breitner, J.C.; Haneuse, S.J.; Walker, R.; Dublin, S.; Crane, P.K.; Gray, S.L.; Larson, E.B. Risk of dementia and AD with prior exposure to NSAIDs in an elderly community-based cohort. Neurology 2009, 72, 1899–1905. [Google Scholar] [CrossRef] [Green Version]
  179. Daniels, M.J.; Rivers-Auty, J.; Schilling, T.; Spencer, N.G.; Watremez, W.; Fasolino, V.; Booth, S.J.; White, C.S.; Baldwin, A.G.; Freeman, S.; et al. Fenamate NSAIDs inhibit the NLRP3 inflammasome and protect against Alzheimer’s disease in rodent models. Nat. Commun. 2016, 7, 12504. [Google Scholar] [CrossRef] [Green Version]
  180. Wang, J.; Tan, L.; Wang, H.F.; Tan, C.C.; Meng, X.F.; Wang, C.; Tang, S.W.; Yu, J.T. Anti-inflammatory drugs and risk of Alzheimer’s disease: An updated systematic review and meta-analysis. J. Alzheimers Dis. 2015, 44, 385–396. [Google Scholar] [CrossRef]
  181. Akinyemi, R.O.; Mukaetova-Ladinska, E.B.; Attems, J.; Ihara, M.; Kalaria, R.N. Vascular risk factors and neurodegeneration in ageing related dementias: Alzheimer’s disease and vascular dementia. Curr. Alzheimer Res. 2013, 10, 642–653. [Google Scholar] [CrossRef] [PubMed]
  182. Sebastiao, I.; Candeias, E.; Santos, M.S.; de Oliveira, C.R.; Moreira, P.I.; Duarte, A.I. Insulin as a bridge between yype 2 diabetes and Alzheimer disease—how anti-diabetics could be a solution for dementia. Front. Endocrinol. (Lausanne) 2014, 5, 110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Fernandez, A.M.; Torres-Aleman, I. The many faces of insulin-like peptide signalling in the brain. Nat. Rev. Neurosci. 2012, 13, 225–239. [Google Scholar] [CrossRef]
  184. Hughes, T.M.; Craft, S. The role of insulin in the vascular contributions to age-related dementia. Biochim. Biophys. Acta 2016, 1862, 983–991. [Google Scholar] [CrossRef] [PubMed]
  185. Barazzoni, R.; Zanetti, M.; Gortan Cappellari, G.; Semolic, A.; Boschelle, M.; Codarin, E.; Pirulli, A.; Cattin, L.; Guarnieri, G. Fatty acids acutely enhance insulin-induced oxidative stress and cause insulin resistance by increasing mitochondrial reactive oxygen species (ROS) generation and nuclear factor-kappaB inhibitor (IkappaB)-nuclear factor-kappaB (NFkappaB) activation in rat muscle, in the absence of mitochondrial dysfunction. Diabetologia 2012, 55, 773–782. [Google Scholar]
  186. Cersosimo, E.; DeFronzo, R.A. Insulin resistance and endothelial dysfunction: The road map to cardiovascular diseases. Diabetes Metab. Res. Rev. 2006, 22, 423–436. [Google Scholar] [CrossRef]
  187. Bhat, N.R. Vasculoprotection as a convergent, multi-targeted mechanism of anti-AD therapeutics and interventions. J. Alzheimers Dis. 2015, 46, 581–591. [Google Scholar] [CrossRef] [Green Version]
  188. Asraf, K.; Torika, N.; Apte, R.N.; Fleisher-Berkovich, S. Microglial activation is modulated by captopril: In vitro and in vivo studies. Front. Cell Neurosci. 2018, 12, 116. [Google Scholar] [CrossRef]
  189. Ongali, B.; Nicolakakis, N.; Tong, X.K.; Aboulkassim, T.; Papadopoulos, P.; Rosa-Neto, P.; Lecrux, C.; Imboden, H.; Hamel, E. Angiotensin II type 1 receptor blocker losartan prevents and rescues cerebrovascular, neuropathological and cognitive deficits in an Alzheimer’s disease model. Neurobiol. Dis. 2014, 68, 126–136. [Google Scholar] [CrossRef]
  190. Anekonda, T.S.; Quinn, J.F. Calcium channel blocking as a therapeutic strategy for Alzheimer’s disease: The case for isradipine. Biochim. Biophys. Acta 2011, 1812, 1584–1590. [Google Scholar] [CrossRef] [Green Version]
  191. Yildirim Simsir, I.; Soyaltin, U.E.; Cetinkalp, S. Glucagon like peptide-1 (GLP-1) likes Alzheimer’s disease. Diabetes Metab. Syndr. 2018, 12, 469–475. [Google Scholar] [CrossRef] [PubMed]
  192. Burns, D.K.; Chiang, C.; Welsh-Bohmer, K.A.; Brannan, S.K.; Culp, M.; O’Neil, J.; Runyan, G.; Harrigan, P.; Plassman, B.L.; Lutz, M.; et al. The TOMMORROW study: Design of an Alzheimer’s disease delay-of-onset clinical trial. Alzheimers Dement. (NY) 2019, 5, 661–670. [Google Scholar] [CrossRef] [PubMed]
  193. Miller, B.W.; Willett, K.C.; Desilets, A.R. Rosiglitazone and pioglitazone for the treatment of Alzheimer’s disease. Ann. Pharmacother. 2011, 45, 1416–1424. [Google Scholar] [CrossRef]
  194. Craft, S.; Baker, L.D.; Montine, T.J.; Minoshima, S.; Watson, G.S.; Claxton, A.; Arbuckle, M.; Callaghan, M.; Tsai, E.; Plymate, S.R.; et al. Intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment: A pilot clinical trial. Arch. Neurol. 2012, 69, 29–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Geifman, N.; Brinton, R.D.; Kennedy, R.E.; Schneider, L.S.; Butte, A.J. Evidence for benefit of statins to modify cognitive decline and risk in Alzheimer’s disease. Alzheimers. Res. Ther. 2017, 9, 10. [Google Scholar] [CrossRef]
  196. Simons, M.; Schwarzler, F.; Lutjohann, D.; von Bergmann, K.; Beyreuther, K.; Dichgans, J.; Wormstall, H.; Hartmann, T.; Schulz, J.B. Treatment with simvastatin in normocholesterolemic patients with Alzheimer’s disease: A 26-week randomized, placebo-controlled, double-blind trial. Ann. Neurol. 2002, 52, 346–350. [Google Scholar] [CrossRef]
  197. Manocha, G.; Ghatak, A.; Puig, K.; Combs, C. Anti-alpha4beta1 integrin antibodies attenuated brain inflammatory changes in a mouse model of Alzheimer’s disease. Curr. Alzheimer Res. 2018, 15, 1123–1135. [Google Scholar] [CrossRef]
  198. Cavalli, G.; Dinarello, C.A. Corrigendum: Anakinra therapy for non-cancer inflammatory diseases. Front. Pharmacol. 2019, 10, 148. [Google Scholar] [CrossRef] [Green Version]
  199. Budni, J.; Bellettini-Santos, T.; Mina, F.; Garcez, M.L.; Zugno, A.I. The involvement of BDNF, NGF and GDNF in aging and Alzheimer’s disease. Aging Dis. 2015, 6, 331–341. [Google Scholar] [CrossRef] [Green Version]
  200. Revilla, S.; Ursulet, S.; Alvarez-Lopez, M.J.; Castro-Freire, M.; Perpina, U.; Garcia-Mesa, Y.; Bortolozzi, A.; Gimenez-Llort, L.; Kaliman, P.; Cristofol, R.; et al. Lenti-GDNF gene therapy protects against Alzheimer’s disease-like neuropathology in 3xTg-AD mice and MC65 cells. CNS Neurosci. Ther. 2014, 20, 961–972. [Google Scholar] [CrossRef]
  201. Harris, R.; Miners, J.S.; Allen, S.; Love, S. VEGFR1 and VEGFR2 in Alzheimer’s Disease. J. Alzheimers Dis. 2018, 61, 741–752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Religa, P.; Cao, R.; Religa, D.; Xue, Y.; Bogdanovic, N.; Westaway, D.; Marti, H.H.; Winblad, B.; Cao, Y. VEGF significantly restores impaired memory behavior in Alzheimer’s mice by improvement of vascular survival. Sci. Rep. 2013, 3, 2053. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Mitra, S.; Behbahani, H.; Eriksdotter, M. Innovative therapy for Alzheimer’s disease-with focus on biodelivery of NGF. Front. Neurosci. 2019, 13, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Nicolakakis, N.; Aboulkassim, T.; Ongali, B.; Lecrux, C.; Fernandes, P.; Rosa-Neto, P.; Tong, X.K.; Hamel, E. Complete rescue of cerebrovascular function in aged Alzheimer’s disease transgenic mice by antioxidants and pioglitazone, a peroxisome proliferator-activated receptor gamma agonist. J. Neurosci. 2008, 28, 9287–9296. [Google Scholar] [CrossRef] [PubMed]
  205. Zolezzi, J.M.; Bastias-Candia, S.; Santos, M.J.; Inestrosa, N.C. Alzheimer’s disease: Relevant molecular and physiopathological events affecting amyloid-beta brain balance and the putative role of PPARs. Front. Aging Neurosci. 2014, 6, 176. [Google Scholar] [CrossRef] [Green Version]
  206. Papadopoulos, P.; Rosa-Neto, P.; Rochford, J.; Hamel, E. Pioglitazone improves reversal learning and exerts mixed cerebrovascular effects in a mouse model of Alzheimer’s disease with combined amyloid-beta and cerebrovascular pathology. PLoS ONE 2013, 8, e68612. [Google Scholar] [CrossRef]
  207. Tong, X.K.; Lecrux, C.; Rosa-Neto, P.; Hamel, E. Age-dependent rescue by simvastatin of Alzheimer’s disease cerebrovascular and memory deficits. J. Neurosci. 2012, 32, 4705–4715. [Google Scholar] [CrossRef] [Green Version]
  208. Silva, I.V.G.; de Figueiredo, R.C.; Rios, D.R.A. Effect of different classes of antihypertensive drugs on endothelial function and inflammation. Int. J. Mol. Sci. 2019, 20, 3458. [Google Scholar] [CrossRef] [Green Version]
  209. Nimmrich, V.; Eckert, A. Calcium channel blockers and dementia. Br. J. Pharmacol. 2013, 169, 1203–1210. [Google Scholar] [CrossRef] [Green Version]
  210. Saavedra, J.M. Angiotensin II AT(1) receptor blockers as treatments for inflammatory brain disorders. Clin. Sci. (Lond.) 2012, 123, 567–590. [Google Scholar] [CrossRef] [Green Version]
  211. Folch, J.; Busquets, O.; Ettcheto, M.; Sanchez-Lopez, E.; Castro-Torres, R.D.; Verdaguer, E.; Garcia, M.L.; Olloquequi, J.; Casadesus, G.; Beas-Zarate, C.; et al. Memantine for the treatment of dementia: A review on its current and future Applications. J. Alzheimers Dis. 2018, 62, 1223–1240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Chen, Z.Z.; Yang, D.D.; Zhao, Z.; Yan, H.; Ji, J.; Sun, X.L. Memantine mediates neuroprotection via regulating neurovascular unit in a mouse model of focal cerebral ischemia. Life Sci. 2016, 150, 8–14. [Google Scholar] [CrossRef] [PubMed]
  213. Abbott, N.J. Astrocyte-endothelial interactions and blood-brain barrier permeability. J. Anat. 2002, 200, 629–638. [Google Scholar] [CrossRef] [PubMed]
  214. Nagelhus, E.A.; Mathiisen, T.M.; Ottersen, O.P. Aquaporin-4 in the central nervous system: Cellular and subcellular distribution and coexpression with KIR4.1. Neuroscience 2004, 129, 905–913. [Google Scholar] [CrossRef] [PubMed]
  215. Wang, X.; Rosell, A.; Lo, E.H. Targeting extracellular matrix proteolysis for hemorrhagic complications of tPA stroke therapy. CNS Neurol. Disord. Drug Targets 2008, 7, 235–242. [Google Scholar] [CrossRef]
  216. Blamire, A.M.; Anthony, D.C.; Rajagopalan, B.; Sibson, N.R.; Perry, V.H.; Styles, P. Interleukin-1beta -induced changes in blood-brain barrier permeability, apparent diffusion coefficient, and cerebral blood volume in the rat brain: A magnetic resonance study. J. Neurosci. 2000, 20, 8153–8159. [Google Scholar] [CrossRef] [Green Version]
  217. Kary, S.; Burmester, G.R. Anakinra: The first interleukin-1 inhibitor in the treatment of rheumatoid arthritis. Int. J. Clin. Pract. 2003, 57, 231–234. [Google Scholar]
  218. Hue, C.D.; Cho, F.S.; Cao, S.; Dale Bass, C.R.; Meaney, D.F.; Morrison, B., 3rd. Dexamethasone potentiates in vitro blood-brain barrier recovery after primary blast injury by glucocorticoid receptor-mediated upregulation of ZO-1 tight junction protein. J. Cereb. Blood Flow Metab. 2015, 35, 1191–1198. [Google Scholar] [CrossRef] [Green Version]
  219. Engelhardt, B.; Kappos, L. Natalizumab: Targeting alpha4-integrins in multiple sclerosis. Neurodegener. Dis. 2008, 5, 16–22. [Google Scholar] [CrossRef] [Green Version]
  220. Bell, R.D.; Zlokovic, B.V. Neurovascular mechanisms and blood-brain barrier disorder in Alzheimer’s disease. Acta Neuropathol. 2009, 118, 103–113. [Google Scholar] [CrossRef] [Green Version]
  221. DiSabato, D.J.; Quan, N.; Godbout, J.P. Neuroinflammation: The devil is in the details. J. Neurochem. 2016, 139, 136–153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Xiong, X.Y.; Liu, L.; Yang, Q.W. Functions and mechanisms of microglia/macrophages in neuroinflammation and neurogenesis after stroke. Prog. Neurobiol. 2016, 142, 23–44. [Google Scholar] [CrossRef] [PubMed]
  223. Zamanian, J.L.; Xu, L.; Foo, L.C.; Nouri, N.; Zhou, L.; Giffard, R.G.; Barres, B.A. Genomic analysis of reactive astrogliosis. J. Neurosci. 2012, 32, 6391–6410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Munch, A.E.; Chung, W.S.; Peterson, T.C.; et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef] [PubMed]
  225. Boado, R.J.; Hui, E.K.; Lu, J.Z.; Zhou, Q.H.; Pardridge, W.M. Selective targeting of a TNFR decoy receptor pharmaceutical to the primate brain as a receptor-specific IgG fusion protein. J. Biotechnol. 2010, 146, 84–91. [Google Scholar] [CrossRef] [Green Version]
  226. Koo, Y.E.; Reddy, G.R.; Bhojani, M.; Schneider, R.; Philbert, M.A.; Rehemtulla, A.; Ross, B.D.; Kopelman, R. Brain cancer diagnosis and therapy with nanoplatforms. Adv. Drug Deliv. Rev. 2006, 58, 1556–1577. [Google Scholar] [CrossRef]
  227. Dadparvar, M.; Wagner, S.; Wien, S.; Kufleitner, J.; Worek, F.; von Briesen, H.; Kreuter, J. HI 6 human serum albumin nanoparticles--development and transport over an in vitro blood-brain barrier model. Toxicol. Lett. 2011, 206, 60–66. [Google Scholar] [CrossRef]
  228. Andrade, S.; Ramalho, M.J.; Loureiro, J.A.; Pereira, M.C. Interaction of natural compounds with biomembrane models: A biophysical approach for the Alzheimer’s disease therapy. Coll. Surf. B Biointerfaces 2019, 180, 83–92. [Google Scholar] [CrossRef]
  229. Surnar, B.; Basu, U.; Banik, B.; Ahmad, A.; Marples, B.; Kolishetti, N.; Dhar, S. Nanotechnology-mediated crossing of two impermeable membranes to modulate the stars of the neurovascular unit for neuroprotection. Proc. Natl. Acad. Sci. USA 2018, 115, E12333–E12342. [Google Scholar] [CrossRef]
  230. Liu, X.; Ye, M.; An, C.; Pan, L.; Ji, L. The effect of cationic albumin-conjugated PEGylated tanshinone IIA nanoparticles on neuronal signal pathways and neuroprotection in cerebral ischemia. Biomaterials 2013, 34, 6893–6905. [Google Scholar] [CrossRef]
  231. Liu, X.; An, C.; Jin, P.; Liu, X.; Wang, L. Protective effects of cationic bovine serum albumin-conjugated PEGylated tanshinone IIA nanoparticles on cerebral ischemia. Biomaterials 2013, 34, 817–830. [Google Scholar] [CrossRef] [PubMed]
  232. Karatas, H.; Aktas, Y.; Gursoy-Ozdemir, Y.; Bodur, E.; Yemisci, M.; Caban, S.; Vural, A.; Pinarbasli, O.; Capan, Y.; Fernandez-Megia, E.; et al. A nanomedicine transports a peptide caspase-3 inhibitor across the blood-brain barrier and provides neuroprotection. J. Neurosci. 2009, 29, 13761–13769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Grammas, P. Neurovascular dysfunction, inflammation and endothelial activation: Implications for the pathogenesis of Alzheimer’s disease. J. Neuroinflamm. 2011, 8, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Ross, C.; Taylor, M.; Fullwood, N.; Allsop, D. Liposome delivery systems for the treatment of Alzheimer’s disease. Int. J. Nanomed. 2018, 13, 8507–8522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Vangilder, R.L.; Rosen, C.L.; Barr, T.L.; Huber, J.D. Targeting the neurovascular unit for treatment of neurological disorders. Pharmacol. Ther. 2011, 130, 239–247. [Google Scholar] [CrossRef] [Green Version]
  236. Joshi, S.; Singh-Moon, R.P.; Ellis, J.A.; Chaudhuri, D.B.; Wang, M.; Reif, R.; Bruce, J.N.; Bigio, I.J.; Straubinger, R.M. Cerebral hypoperfusion-assisted intra-arterial deposition of liposomes in normal and glioma-bearing rats. Neurosurgery 2015, 76, 92–100. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Illustration of the constitutive cellular elements of the neurovascular unit (NVU) along the entire brain vasculature. Panel (a) shows a panoramic view of the vascular tree. Panels (be) depict the different levels of the vascular tree. Pial arteries (panel (b)) run along the brain surface and penetrate the parenchyma, narrowing and branching into penetrating arterioles (panel (c)), which form the intraparenchymal arterioles (panel (d)), and ultimately give rise to capillaries (panel (e)). Abbreviations: IEL, internal elastic lamina; PVM, perivascular macrophage; SAS, the subarachnoid space; SMC, smooth muscle cell.
Figure 1. Illustration of the constitutive cellular elements of the neurovascular unit (NVU) along the entire brain vasculature. Panel (a) shows a panoramic view of the vascular tree. Panels (be) depict the different levels of the vascular tree. Pial arteries (panel (b)) run along the brain surface and penetrate the parenchyma, narrowing and branching into penetrating arterioles (panel (c)), which form the intraparenchymal arterioles (panel (d)), and ultimately give rise to capillaries (panel (e)). Abbreviations: IEL, internal elastic lamina; PVM, perivascular macrophage; SAS, the subarachnoid space; SMC, smooth muscle cell.
Ijms 22 02022 g001
Figure 2. Schematic representation of the NVU under physiological (a) and pathological (b) conditions. Abbreviations: NFT, neurofibrillary tangle; NP, neuritic plaque.
Figure 2. Schematic representation of the NVU under physiological (a) and pathological (b) conditions. Abbreviations: NFT, neurofibrillary tangle; NP, neuritic plaque.
Ijms 22 02022 g002
Figure 3. Overview of the complex cell–cell signaling in the NVU, triggered by extracellular and vascular Aβ deposits. Step 1 shows the interaction of Aβ oligomers and fibrils with NVU cells through several receptors. Step 2 illustrates the altered Aβ clearance that triggers NVU dysfunction. Step 3 shows the NVU cell dysfunction as the causative agent of cerebral hypoxia. Step 4 shows cerebral hypoxia, neuroinflammatory environment, and peripheral blood infiltrate as the promoting causes of neuronal degeneration. Abbreviations: AQ-4, aquaporin-4; Aβ, amyloid peptide; BBB, blood-brain barrier; CBF, cerebral blood flow; GLUT, glucose transporter; LRP1, low-density lipoprotein receptor-related protein 1; MEOX2, mesenchyme homeobox 2; MMP, matrix metalloproteinase; MYOCD, myocardin; NFκB, nuclear factor kappa light chain enhancer of activated B cells; PDGF-B, platelet-derived growth factor subunit B; PDGFRβ, platelet-derived growth factor receptor β; PGE2, prostaglandin E2; TGFβ, transforming growth factor-beta; RAGE, receptor for advanced glycation end products; ROS, reactive oxygen species.
Figure 3. Overview of the complex cell–cell signaling in the NVU, triggered by extracellular and vascular Aβ deposits. Step 1 shows the interaction of Aβ oligomers and fibrils with NVU cells through several receptors. Step 2 illustrates the altered Aβ clearance that triggers NVU dysfunction. Step 3 shows the NVU cell dysfunction as the causative agent of cerebral hypoxia. Step 4 shows cerebral hypoxia, neuroinflammatory environment, and peripheral blood infiltrate as the promoting causes of neuronal degeneration. Abbreviations: AQ-4, aquaporin-4; Aβ, amyloid peptide; BBB, blood-brain barrier; CBF, cerebral blood flow; GLUT, glucose transporter; LRP1, low-density lipoprotein receptor-related protein 1; MEOX2, mesenchyme homeobox 2; MMP, matrix metalloproteinase; MYOCD, myocardin; NFκB, nuclear factor kappa light chain enhancer of activated B cells; PDGF-B, platelet-derived growth factor subunit B; PDGFRβ, platelet-derived growth factor receptor β; PGE2, prostaglandin E2; TGFβ, transforming growth factor-beta; RAGE, receptor for advanced glycation end products; ROS, reactive oxygen species.
Ijms 22 02022 g003
Table 1. Interaction among different species of Aβ aggregates (f: fibrillar; o: oligomeric; m: monomeric; *: unknown), receptors in NVU cells, and pathological effects.
Table 1. Interaction among different species of Aβ aggregates (f: fibrillar; o: oligomeric; m: monomeric; *: unknown), receptors in NVU cells, and pathological effects.
Aβ Receptor and LigandNVU CellsPathological EffectsRef.
CR1 (CD35) fAβMicroglia, astrocytes, neurons fAβ/CR1 interaction results in C3b/C4b activation. Aβ clearance from the brain via blood cell expresses CR1 in its surface and later metabolism in the liver and/or spleen.[70,71]
CR3 (Mac-1) fAβMicroglia Interaction leads to an increased PI3K/p47PHOX activity (neurotoxicity by superoxide) or NF-kB (inflammatory factors production). [72,73]
TLR4/6 fAβMicroglia, astrocytesCD36/TLR4/6 complex mediates Aβ internalization, followed by ROS and proinflammatory production and phagocytosis.[74,75]
C5aR (CD88) fAβ, oAβC5a/C5aR binding in response to fAβ/oAβ induces TNFα production. [76,77]
SRA 1/2
fAβ, oAβ
Aβ/SRA interaction results in NF-kB activation and consequently, the secretion of ROS, TNF-a, complement components, among other pro-inflammatory substances. [78,79]
SRB2 (CD36) fAβ, oAβMicroglia, BECs, neuronsCD36/a3b1-integrin/CD47 complex regulates fAβ interaction in microglia cells and triggers ROS production, pro-inflammatory cytokines release, and phagocytosis. [79,80]
RAGE mAβ, fAβAβ/RAGE/p38 and ERK1/2 signaling pathways trigger oxidative stress, NF-kB activation, proinflammatory molecules production, triggering NVU damage. [81,82]
a7nAChR mAβ, fAβNeurons, SMCs, astrocytesa7nAChR may mediate Aβ internalization. Aβ could activate the JNK/ERK2/MAPK pathway, which results in cell death by apoptosis.[83,84]
IR mAβ, oAβneuronsAβ/IR binding triggers impaired insulin signaling, which could cause neuronal dysfunction and memory deficits.[85,86]
SEC-R mAβNeurons, gliaInteraction mediates endocytosis and degradation of Aβ by recognizing its 25–35 region. [87,88]
TREM2 oAβMicroglia, neuronsDecreased TREM2 leads to Aβ accumulation. TREM2/Aβ linking could trigger neuronal phagocytosis or apoptosis.[89,90]
LRP1 mAβPericytes, astrocyte, microglia, neuronsLRP1 is widely expressed in NVU cells and mediates the Aβ transport across the BBB. LRP1 controls the Aβ uptake and its subsequent trafficking to the lysosome for degradation. [91,92]
ABCA1 * AβBECs, pericytesABCA1/ApoE/LRP1 complex contributes to brain Aβ transport/clearance. Abca1 gene deficiency promotes Aβ accumulation in an AD mice model.[93]
ABCB1 * AβABCB1/LRP1 transports the Aβ peptides across the BBB. ABCB1 is considered a marker for BBB maturity and functionality.[94]
Abbreviations: ABCA1, ATP binding cassette A1; ABCB1, ATP-binding cassette sub-family B member 1; ApoE, Apolipoprotein E; BBB, blood-brain barrier; BECs, brain endothelial cells; CR, complement receptor; Cx, complement receptor; fAβ, filamentous Aβ; IR, insulin receptor; LRP1, low-density lipoprotein receptor-related protein 1; NF-kB, nuclear factor-ΚB; NVU, neurovascular unit; oAβ, oligomeric Aβ; p47PHOX, neutrophil cytosol factor 1; PI3K, phosphatidylinositol 3-kinase; ROS, reactive oxygen species; SEC-R, serpin-enzyme complex receptor; SMCs, smooth muscle cells; SRA, Class A scavenger receptor; SRB2, scavenger receptor class B member 2; TLR, Toll-like receptor; TREM2, triggering receptor expressed on myeloid cells 2.
Table 2. The effects of potential AD pharmacological therapies on the NVU.
Table 2. The effects of potential AD pharmacological therapies on the NVU.
Drug/CT IdentifierClinical PhaseEffects on the NVU
(a) BACE 1 inhibitors
E2609/NCT01294540 ¥IEffect on the neuron and BEC. They reduce extracellular Aβ accumulation. Inhibition of the endothelial BACE1 activity may reduce Aβ release into the SMCs and protect vascular wall integrity [117].[118]
Verubecestat/NCT01739348 ¥III[119]
Umibecestat/NCT01097096 ¥III[120]
LY2886721/NCT01561430 ¥III[121]
(b) Gamma-secretase inhibitors and modulators
Semagacestat/NCT00594568 ¥IIIEffect on NVU cells. They block Aβ-peptide secretion, precursor agent of NPs, and neuronal dysfunction. According to an animal AD model, some of these drugs can interact with astrocytes and microglia, preventing the Aβ secretion and decreasing the total brain amyloid load [122].[123]
Avagacestat/NCT00810147 ¥II[124]
Pintol/NCT00470418 *II[121]
(c) Alpha-secretase enhancers
Epigallocatechin gallate/NCT00951834, NCT03978052 ΦII/IIIEffect on NVU cells. Some of these drugs have been shown to improve NVU in animal models, reducing vascular Aβ deposits, neuroinflammation, oxidative stress, synaptotoxicity, and favoring angiogenesis and antioxidant and anti-inflammatory effects [125,126]. [121]
Bryostatin 1/NCT04538066 ΦII[127]
Etazolate/NCT00880412 *II[128]
(d) β-Amyloid aggregation inhibitors
Scyllo-inositol (ELND005) ¥IIEffect on NVU cells. The Aβ polymerization process is a key event involved in AD. Some of these drugs could inhibit Aβ self-association [129]. Therefore, they could inhibit vascular insoluble amyloid deposits and improve the functionality of the NVU in AD.[130]
Clioquinol (PBT1) &III[121]
Colostrinin *III[121]
(e) Modulators of beta-amyloid peptide transport-RAGE inhibitors
Azeliragon (PF-04494700)/NCT03980730 ΦIIEffect on microglia, BEC, and neurons. They regulate the Aβ clearance and improve the BBB functionality, neuroinflammatory environment, and CBF.[131]
TTP4000/NCT01548430 *I[132]
(f) Tau aggregation inhibitors
Rember/NCT00515333 *IIEffect on the neuron. Effects on other NVU cells unknown. They prevent oligomeric tau aggregation and NFT formation and disrupt aggregated tau. [133]
LMTM/NCT01689246, NCT03539380, NCT03446001 ΦIII[133]
(g) Kinases inhibitors
Tideglusib (GSK3β inhibitor)/NCT00948259, NCT01350362 *IIIEffect on the neuron. No effect on other NVU cells. Inhibition of kinases leads to tau phosphorylation, loss of affinity for microtubules, NFT formation, and neuronal death.[133]
Saracatinib (Fyn inhibitor)/NCT01864655, NCT02167256 *II[133]
Nilotinib (tyrosine kinase inhibitor)/NCT02947893ΦII[134]
(h) Microtubule Stabilizers
Epothilone D/NCT01492374 ¥IEffect on the neuron. No impact on other NVU cells. They stabilize microtubules and reduce tau pathology and hippocampal neuronal loss.[133]
Abetotexate/NCT01966666 *I[133]
(i) Active immunotherapy anti-tau protein
AADvac-1/NCT01850238 *IEffect on the neuron. No effect on other NVU cells. It activates T cells and triggers an immune response to eliminate abnormally phosphorylated tau. [133]
(j) Active immunity (anti-Aβ peptide polyclonal antibody)
Vanutide cridificar/NCT00479557, NCT01227564 *IIaEffect on the neuron. It induces antibodies against Aβ, preventing Aβ deposition, promoting plaque clearance, and improving cognitive functions.[135]
(k) Passive immunotherapy (anti-tau antibody)
R07105705 (RG6100)/NCT02820896, NCT0328914 ΦIIEffect on the neuron. No effect on other NVU cells. It recognizes abnormal tau and blocks its transmission from one neuron to another. [133]
(l) Passive immunity (anti-Aβ monoclonal antibodies)
Aducanumab (IgG1)/NCT04241068 ΦIIIEffect on the neuron and possibly on other cells of the NVU. Most of these drugs are directed against epitopes of aggregated forms of Aβ, including soluble monomers, oligomers, and insoluble fibrils. Besides, they favor the central Aβ clearance (except Ponezumab that promotes peripheral release), having a favorable effect on neurons and the other NVU cells. Donanemab, unlike the others, has an affinity for the Aβp3-42 conformation, which is more toxic than Aβ1-40 or 1–42 [136,137]. Furthermore, IgG1 or IgG2 carries a high risk of Fc γ receptor (FcγRs)-mediated overactivation of microglial cells, due to the binding with C1q that can contribute to an inappropriate pro-inflammatory response leading to vasogenic edema and cerebral microbleeds, an effect that does not occur with IgG4. In general, both active and passive immunotherapy tend to have harmful effects on the NVU, increasing the CAA and causing microhemorrhages [138].[139]
Gantenerumab (IgG1)/NCT01760005 ΦIII[121]
Donanemab (IgG1)/NCT03367403ΦII[140]
Solanezumab (IgG)/NCT00905372, NCT01760005 ΦIII[141]
Crenezumab (IgG4)/NCT03977584, NCT01998841 ΦII[142]
Sar228810 (IgG4)/NCT01485302 *I[143]
GSK933776A (IgG1)/NCT00459550 *I[144]
Ponezumab (IgG2)/
NCT00722046, NCT00945672 *
II[145]
BAN-2401 (IgG1)/NLT01767311 NCT03887455, NCT04468659 ΦIIb[146]
(m) Passive immunity (Intravenous immunoglobulin G)
Octagam IVIgG/NCT01300728 ΦIIIEffects on the NVU cells. They promote central Aβ clearance, block the RAGE receptor, increases sLRP levels, anti-inflammatory effects, and selectively target aggregated Aβ forms (monomers and oligomers).[147]
Gammagard IVIgG/NCT00818662 *III[147]
(n) Nonsteroidal anti-inflammatory drugs (NSAIDs)
r-Flurbiprofen ζIIIEffects on the NVU cells. γ-secretase inhibitor acts restoring neurogenesis, reorganizing the astrocytic cytoskeleton, reducing pathological tau, rescuing synaptic plasticity, acting on microglia to counteract neuroinflammation[148]
Itanapraced/NCT01303744 ζII[121,149]
¥ Suspended due to adverse effects. * All studies completed but no current active study. Φ Ongoing clinical study. & Suspended for toxic environmental effects. ζ Suspended due to financing/licensing issues. Abbreviations: Aβ, amyloid β-peptide; ACE, angiotensin-converting enzyme; AD, Alzheimer’s disease; APP, amyloid precursor protein; sAPPα, secreted ectodomain APP alpha; ARB, angiotensin receptor blocker; BACE1, beta-site APP cleaving enzyme 1; BBB, blood-brain barrier; BECs, brain endothelial cells; C1q, complement factor; CBF, cerebral blood flow; eNOS, endothelial nitric oxide synthase; Fyn, proto-oncogene tyrosine-protein kinase Fyn; GDNF, glial cell-derived neurotrophic factor; GLP-1, glucagon-like peptide 1; GSK3β, glycogen synthase kinase 3β; HMG-CoA, 3-hydroxy-3-methylglutaryl-CoA; IgG, immunoglobulin G; IL-1β, interleukin-1β; IL-1R1, interleukin 1 receptor type 1; NFTs, neurofibrillary tangles; NPs, neuritic plaques; NVU, neurovascular unit; Pcp, preclinical phase; RAGE, receptor for advanced glycation endproducts; sLRP, soluble low-density lipoprotein receptor-related protein; SMCs, smooth muscle cells; VEGF, vascular endothelial growth factor; γ-PPAR, peroxisome proliferator-activated receptor-γ.
Table 3. The effects of potential therapies focused on preventing damage to or restoring function of the NVU in AD.
Table 3. The effects of potential therapies focused on preventing damage to or restoring function of the NVU in AD.
Drug/CT IdentifierClinical PhaseEffects on the NVURef
(a) Antihypertensive drugs
Captopril (ACE inhibitor)PcpEffects on the NVU cells. It inhibits Aβ production caused by ACE, regulates pro-inflammatory molecules, and inhibits ROS.[188]
Losartán (ARBs)/NCT02913664 ΦIIEffects on the NVU cells. It inhibits ROS production and reduces cerebrovascular/neuropathological changes.[189]
Amlodipine/NCT02913664 Φ Nilvadipine/NCT02017340 * (Calcium-channel blockers).II IIIEffects on the NVU cells. They attenuate the neuronal deterioration induced by Aβ, decrease cerebral hypoperfusion due to vasodilator effects.[190]
(b) Antidiabetic drugs
Exenatide/NCT01255163 ζ Liraglutide/NCT01843075 Φ (GLP-1 agonists)IINeuron and microglia effects. They promote neuronal survival, synaptogenesis, neurogenesis, anti-inflammation, and protecting against oxidative injury. In AD mouse models reduce Aβ oligomers and plaque load, and microglial activation, improving memory. Also, elicit vasculoprotective effects (protect against hypoxia injury).[191]
Pioglitazone/NCT01456117 NCT00982202 (γ-PPAR)
Rosiglitazone/NCT00550420 NCT00265148 (γ-PPAR) *
IIIEffects on the NVU cells. They reduce Aβ levels (enhanced phagocytosis of Aβ deposits), oxidative stress, mitochondrial dysfunction, and neuroinflammation induced by glial cells. They improve cerebral blood flow, and lead to cognitive improvement. [192]
[193]
Intranasal insulin/NCT00438568 NCT01547169, NCT01436045 *IINeuron effects. They modulate the Aβ levels, protect against synapses damage by Aβ oligomers, and modulate memory consolidation.[194]
Simvastatine (HMG-CoA)/NCT00053599, NCT00939822, NCT00486044, NCT00303277 *IIEffects on the NVU cells. They have neuroprotective and pleiotropic impact, improve the vascular system, eNOS activation, and antioxidant effects.[195,196]
(c) Novel drugs
Natalizumab (antibodies against the α4β1 integrin receptor)PcpEffects on the NVU cells. It acts modulating the peripheral immune system infiltrate into the brain and decrease the proinflammatory environment. [197]
Anakinra (IL-1 receptor antagonist)PcpProbable effects on microglia cells. It blocks the interaction between IL-1β with its receptor (IL-1R1), then could decrease neuroinflammation in AD patients.[198]
GDNF/lentiviral vectorPcpIt has neuroprotector effects against Aβ, protecting neurons and astrocytes. [199,200]
VEGFPcpEffects on the NVU cells. It modulates angiogenesis, vascular permeability, vascular remodeling, vascular survival, neurotrophic activity, and anti-inflammation.[201,202]
NGF and NGF mimetics/NCT03069014 *(Intracerebral infusion, nasal/intraocular administration)IIEffects on the neuron and microglia. They regulate differentiation, growth, survival, and plasticity of cholinergic neurons. They have a direct role in modulating microglial cells toward a non-inflammatory phenotype.[203]
Φ Ongoing clinical study. * All studies completed but no current active study. ζ Suspended due to financing/licensing issues. Suspended for lack of effectiveness. Abbreviations: Aβ, amyloid β-peptide; ACE, angiotensin-converting enzyme; AD, Alzheimer’s disease; C.p., clinical phase; eNOS, endothelial nitric oxide synthase; Fyn, proto-oncogene tyrosine-protein kinase Fyn; GDNF, glial cell-derived neurotrophic factor; GLP-1, glucagon-like peptide 1; GSK3β, glycogen synthase kinase 3β; HMG-CoA, 3-hydroxy-3-methylglutaryl-CoA; IgG, immunoglobulin G; IL-1β, interleukin-1β; IL-1R1, interleukin 1 receptor type 1; NFTs, neurofibrillary tangles; NGF, nerve growth factor; NPs, neuritic plaques; NVU, neurovascular unit; Pcp, preclinical phase; ROS, reactive oxygen species; sLRP, soluble low-density lipoprotein receptor-related protein; SMCs, smooth muscle cells; VEGF, vascular endothelial growth factor; γ-PPAR, peroxisome proliferator-activated receptor-γ.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Soto-Rojas, L.O.; Pacheco-Herrero, M.; Martínez-Gómez, P.A.; Campa-Córdoba, B.B.; Apátiga-Pérez, R.; Villegas-Rojas, M.M.; Harrington, C.R.; de la Cruz, F.; Garcés-Ramírez, L.; Luna-Muñoz, J. The Neurovascular Unit Dysfunction in Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 2022. https://doi.org/10.3390/ijms22042022

AMA Style

Soto-Rojas LO, Pacheco-Herrero M, Martínez-Gómez PA, Campa-Córdoba BB, Apátiga-Pérez R, Villegas-Rojas MM, Harrington CR, de la Cruz F, Garcés-Ramírez L, Luna-Muñoz J. The Neurovascular Unit Dysfunction in Alzheimer’s Disease. International Journal of Molecular Sciences. 2021; 22(4):2022. https://doi.org/10.3390/ijms22042022

Chicago/Turabian Style

Soto-Rojas, Luis O., Mar Pacheco-Herrero, Paola A. Martínez-Gómez, B. Berenice Campa-Córdoba, Ricardo Apátiga-Pérez, Marcos M. Villegas-Rojas, Charles R. Harrington, Fidel de la Cruz, Linda Garcés-Ramírez, and José Luna-Muñoz. 2021. "The Neurovascular Unit Dysfunction in Alzheimer’s Disease" International Journal of Molecular Sciences 22, no. 4: 2022. https://doi.org/10.3390/ijms22042022

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop