IMR Press / FBL / Volume 29 / Issue 4 / DOI: 10.31083/j.fbl2904136
Open Access Review
Interactions between Beta-Amyloid and Pericytes in Alzheimer’s Disease
Show Less
1 Department of Neurology, Qilu Hospital of Shandong University, 250012 Jinan, Shandong, China
2 The Second Clinical Medical School of Shandong University, 250033 Jinan, Shandong, China
*Correspondence: nancyduan94@hotmail.com (Ruo-Nan Duan); yili202@hotmail.com (Yi Li)
Front. Biosci. (Landmark Ed) 2024, 29(4), 136; https://doi.org/10.31083/j.fbl2904136
Submitted: 29 October 2023 | Revised: 20 January 2024 | Accepted: 25 January 2024 | Published: 2 April 2024
Copyright: © 2024 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Alzheimer’s disease (AD) is an age-related progressive neurodegenerative disorder characterized by aberrant amyloid precursor protein (APP) cleavage, pathological aggregations of beta-amyloid (Aβ) that make up Aβ plaques and hyperphosphorylation of Tau that makes up neurofibrillary tangles (NFTs). Although progress has been made in research on AD, the fundamental causes of this disease have not been fully elucidated. Recent studies have shown that vascular dysfunction especially the loss of pericytes plays a significant role in the onset of AD. Pericytes play a variety of important roles in the nervous system including the regulation of the cerebral blood flow (CBF), the formation and maintenance of the blood–brain barrier (BBB), angiogenesis, and the clearance of toxic substances from the brain. Pericytes participate in the transport of Aβ through various receptors, and Aβ acts on pericytes to cause them to constrict, detach, and die. The loss of pericytes elevates the levels of Aβ1-40 and Aβ1-42 by disrupting the integrity of the BBB and reducing the clearance of soluble Aβ from the brain interstitial fluid. The aggravated deposition of Aβ further exacerbates pericyte dysfunction, forming a vicious cycle. The combined influence of these factors eventually results in the loss of neurons and cognitive decline. Further exploration of the interactions between pericytes and Aβ is beneficial for understanding AD and could lead to the identification of new therapeutic targets for the prevention and treatment of AD. In this review, we explore the characterization of pericytes, interactions between pericytes and other cells in the neurovascular unit (NVU), and the physiological functions of pericytes and dysfunctions in AD. This review discusses the interactions between pericytes and Aβ, as well as current and further strategies for preventing or treating AD targeting pericytes.

Keywords
Alzheimer's disease
pericytes
beta-amyloid
cerebral blood flow
blood-brain barrier
neurovascular unit
cognitive decline
1. Introduction

Alzheimer’s disease (AD) is an age-related progressive neurodegenerative disorder characterized by cognitive dysfunction and behavioral impairment. In addition, AD is the primary cause of dementia [1]. The two typical neuropathological changes in AD include neuritic plaques formed by the deposition of beta-amyloid (Aβ) in the brain parenchyma and intracellular Tau that makes up neurofibrillary tangles (NFTs) formed by the accumulation of hyperphosphorylated tau proteins [2, 3].

The plaques of Aβ also deposit on vessel walls causing cerebral amyloid angiogenesis (CAA) [4, 5]. However, the Aβ plaques in AD and CAA are distinct. Aβ1-40 and Aβ1-42 are the most common subtypes of Aβ peptide. Aβ1-42 is more prone to form insoluble aggregates in the parenchyma [6], constituting the major component of neuritic plaques in AD. In contrast, Aβ1-40 aggregates more slowly and ultimately deposits in the walls of vessels through perivascular drainage [4, 7, 8]. Abnormal perivascular drainage is the main pathogenesis of CAA [4, 9, 10], which also explains why the Aβ deposited on the vascular wall in CAA is mainly Aβ1-40. Neuron loss and synapse dysfunction caused by the toxicity of Aβ will ultimately contribute to dementia and degeneration of the central nervous system (CNS) [11, 12]. Moreover, a substantial body of research has demonstrated that the occurrence of CAA and AD largely overlap [13, 14, 15]. Cerebrovascular dysfunction caused by CAA is related to severe cognitive impairment in AD patients [15, 16, 17, 18].

The shared role of Aβ in AD and CAA is likely the most apparent interaction between neurodegenerative diseases and cerebrovascular diseases. Moreover, there is evidence indicating that vascular dysfunction plays a significant role in the onset of AD. Recent studies have demonstrated that a reduction in the cerebral blood flow (CBF) is the earliest detectable clinical change in mild cognitive impairment and AD patients [19, 20, 21] and that capillaries exhibit focal constriction. The greatest vascular resistance occurs in the capillary bed rather than in the penetrating arterioles [22]. At the capillary level, the neurovascular unit (NVU) is composed of endothelial cells (ECs), pericytes, glial cells and neurons. Pericytes are the only contractile cells responsible for regulating blood flow in capillaries. Pericytes likely plays an important role in the pathogenesis of AD. Pericytes, which are indispensable components of the NVU, play an essential role in the formation and maintenance of the blood-brain barrier (BBB), the regulation of the CBF, angiogenesis and the phagocytosis of toxic substances including Aβ from the brain. A significant loss of pericytes in AD patients has been observed, and the accumulation of Aβ may be the potential cause. Conversely, the loss of pericytes could lead to impaired clearance of Aβ, exacerbating the deposition of Aβ and leading to a vicious cycle.

Currently, there are no effective drugs that can effectively reverse cognitive decline and there are no therapeutic strategies targeting pericytes [23]. Further understanding of the pathological changes in pericytes in AD and the interactions between pericytes and Aβ may provide new therapeutic directions for the prevention and treatment of AD. In this review, we summarize the characterization of pericytes, the signaling pathways linking pericytes and other cells in the NVU, the physiological effects of pericytes, the functional changes in pericytes in AD, the pathways through which pericytes clear Aβ, the effects of Aβ on pericytes and the current strategies for preventing or treating AD targeting pericytes.

2. The Characterization of Pericytes

Pericytes were originally characterized by Eberth and Rouget in the 1870s (Eberth, 1871; Rouget, 1873) and firstly named by Zimmermann in 1923 based on their location within the vascular basement membrane (BM) and the extension of cytoplasmic processes to wrap ECs. Both pericytes and vascular smooth muscle cells (VSMCs) are called mural cells [24]. In addition to ring-shaped VSMCs with circumferential processes on arteries and arterioles [25], pericytes are classified into three subtypes based on their morphology and location: ensheathing pericytes which have more circumferential processes on precapillary arterioles; thin-strand or helical pericytes, which have protruding nuclei and longitudinal processes on the middle capillary, which is the most widely accepted morphology of pericytes; and stellated pericytes on the postcapillary space [26, 27].

Many cell surface proteins such as platelet-derived growth factor receptor-β (PDGFR-β), neural/glial antigen 2 (NG2) and CD13 [28, 29, 30, 31, 32, 33] are expressed on both pericytes and VSMCs, and these two cell types can be distinguished by morphology. Additionally, vitronectin (VTN) and interferon-induced transmembrane protein 1 (Ifitm-1) label pericytes specifically [31]. However, there is currently a lack of specific markers for distinguishing subpopulations of pericytes. Pericytes and VSMCs exhibit contractile alpha-smooth muscle actin (α-SMA) and desmin expression [26, 30, 34]. Notably, there is a difference in the expression level of α-SMA between the subtypes of pericytes, which may be related to their distinct functions [27, 30]. These markers, especially PDGFR-β, NG2 and α-SMA, are widely applied in studies. PDGFR-β can outline the contours of pericytes [35]. Because they are labeled by PDGFR-β, pericytes are easily recognized by their protruding soma. Therefore, relying solely on morphology is sufficient to reliably identify pericytes [26, 27]. However, it is worth noting that adequate experience is needed for observers [36]. NG2 is the first discovered marker of pericytes that can be used to identify pericytes through combination with morphology, but not all pericyte subsets express NG2 [37]. α-SMA is not sensitive enough to identify pericytes in capillary beds, because pericytes on precapillary tubes express more α-SMA while pericytes on capillary beds may be negative [27, 30].

3. The Interactions between Pericytes and other Cells in the Neurovascular Unit

The NVU is composed of endothelial cells, mural cells (vascular smooth muscle cells, pericytes), glial cells (astrocytes, microglia, oligodendrocytes) and neurons [38, 39, 40]. The cellular components vary with the branching of the cerebral vascular tree. At the capillary level, pericytes are located centrally between endothelial cells, the endfeet of pericytes and neurons, and the BM is shared with pericytes [26, 41]. They communicate with their neighboring cells and generate corresponding responses which are crucial for normal functions of the CNS [26]. We reviewed the interactions between pericytes and ECs, astrocytes and neurons in Table 1 (Ref. [26, 35, 38, 42, 43, 44, 45, 46, 47, 48, 49, 50]).

Table 1.The interactions between pericytes and other cells in the NVU.
Cell type Signaling pathway with pericytes Functions Ref
ECs PDGF-BB-PDGFRβ pathway PDGF-BB secreted by ECs combines with PDGFRβ on pericytes in high affinity. PDGF-BB-PDGFRβ signaling promotes pericytes survival, proliferation and migration. [26, 35, 42]
TGF-β–TGFβR2 pathway TGF-β is activated though interactions between ECs and pericytes to promote proliferation and differentiation of pericytes, and stabilization of vessels. [43, 44]
Ang-Tie2 pathway Ang1 secreted by pericytes combines with Tie2 on ECs. Ang-Tie2 signaling regulate angiogenesis and vascular permeability. [26, 35, 45]
VEGF-A-VEGFR2 pathway VEGF-A secreted by pericytes and ECs to promote the survival and proliferation of pericytes as well as angiogenesis. [26, 38, 46]
Astrocytes CypA–NFκB–MMP-9 pathway ApoE secreted by astrocytes interacts with LRP1 on pericytes triggering the degradation of the extracellular matrix and tight junction. [26, 47, 48]
Pericytes regulate the AQP4 distribution of astrocytes to regulate the polarization of astrocytic endfeet. [49, 50]
Neurons Pericytes secrete neurotrophic factors to promote the survival of neurons whereas neurons secrete neurotransmitters to regulate pericytes contractility. [26, 50]

Abbreviations: NVU, neurovascular unit; EC, endothelial cells; PDGF-BB, platelet-derived growth factor-BB; PDGFRβ, platelet-derived growth factor receptor-β; TGF-β, transforming growth factor-β; TGFβR2, transforming growth factor-β receptor 2; Ang1, angiopoietin-1; Tie2, tyrosine protein kinase receptor; VEGF-A, vascular endothelial growth factor-A; VEGFR2, vascular endothelial growth factor receptor 2; CypA, cyclophilin A; NFκB, nuclear factor kappa-B; MMP-9, matrix metalloproteinase-9; ApoE, apolipoprotein E; LRP1, LDL receptor-related protein-1; AQP4, aquaporin 4.

4. The Functions of Pericytes and Dysfunctions in AD

As pericytes are indispensable components of the BBB and NVU, we review the roles of pericytes in the CNS and their dysfunctions in AD.

4.1 Regulation of the Cerebral Blood Flow (CBF)

Mural cells are cellular components with contractile properties in the NVU, that enable them to regulate vascular tone and the CBF [22]. As pial arteries branch into arterioles and capillaries after penetrating into parenchyma, the mural cell population composed of the NVU changes [51]. Penetrating arteries consist of one to three layers of VSMCs while arterioles contain only one layer [41, 51]. After descending to capillary level, pericytes replace VSMCs and embed within the endothelial BM [51]. Previously, the CBF was shown to be regulated solely by VSMCs [52]. However, with the study of pericytes, this viewpoint has been challenged.

In a series of studies, pericyte were confirmed to constrict or dilate in response to neurotransmitters [53], for example, glutamate evokes pericyte dilation, and pericytes constrict in response to a gamma-aminobutyric acid (GABA) receptor blocker suggesting that pericytes participate in the regulation of the CBF [52, 54, 55, 56, 57]. Moreover, an in vivo experiment in which mice expressed DsRed in pericytes, revealed that capillary dilation precedes penetrating arterioles, demonstrating that capillary dilation is a result of active relaxation of pericytes rather than a passive response to elevated blood pressure caused by arteriole dilation [52]. In ischemic stroke, capillaries constrict segmentally at regions near pericytes, after which pericytes contract and subsequently die rigidly [58]. Damages to pericytes contributes to long-lasting microcirculatory reflow impairment even after reperfusion [59].

Pericytes degeneration and neurovascular dysfunction have been observed in AD [51, 52]. Moreover, the oxidative stress caused by Aβ leads to capillary constriction. A reduction in the CBF reduces the oxygen supply and glucose availability to the brain, resulting in the impairment of neurons and neurodegenerative changes [60]. A recent study showed that white matter lesions (WMLs) induced by persistent cerebral hypoperfusion is a driving factor for dementia [61]. Moreover, the two-hit vascular hypothesis suggests that prior to neurodegeneration and cognitive impairment, genetic, vascular and environmental factors cause vascular damage (hit1), and neurovascular dysfunction contributes to the accumulation of Aβ (hit 2) [40, 41]. A previous study using APPsw/0 Pdgfrβ+/- mice found that the loss of pericytes results in a series of AD-like neurodegeneration pathological changes including accelerated Aβ deposition, tau pathology and neuronal dysfunction [62]. Notably, vascular damage (hit1) has been observed in pericyte-deficient APPsw/0 mice, indicating that vessel damage and pericyte degeneration may be mutually causal [62]. Together, these findings suggest that pericyte degeneration is an early and key event in AD neurodegeneration.

However, due to differences in stimulation methods, transgenic mice, and other factors, the function of pericytes in regulating the CBF has not been fully elucidated.

4.2 Formation and Maintenance of the Blood-Brain Barrier (BBB)

The blood-brain barrier (BBB) is a special protective barrier that exists between capillaries and the brain, and is composed of ECs, endothelial tight junctions (TJs), the BM, pericytes and astrocyte endfeet [63, 64]. The BBB protects the brain from invasion by blood-derived harmful factors, thus maintaining the homeostasis of the CNS [65]. Using quail-chick transplantation chimeras, BBB was shown to develop in response to the neural tissue environment [66]. During embryogenesis in rodents, astrocytes and pericytes are required to wrap immature vessels. It is widely accepted that immature vessels are covered preferentially by astrocytes postnatally. However, Daneman et al. [67] reported that pericytes were recruited during embryogenesis, more than one week before the generation of astrocytes, thus revealing the role of pericytes in the formation of the BBB. The permeability of the BBB is increased in pericyte-deficient mice, indicating the essential role of pericytes in maintaining BBB integrity [49]. Pericytes maintain the integrity of the BBB through two pathways, forming and preserving the TJs of ECs, and transcytosing in the CNS ECs.

The breakdown of the BBB has been observed in AD [68], which is attributed to the loss and detachment of pericytes destroying the integral structure of the BBB and increasing BBB permeability [69]. Interestingly, sirtuin-1 (SIRT1), an anti-aging gene, is markedly suppressed exposed to Aβ [70]. The decreased expression of SIRT1 also increased the permeability of the BBB and accelerated the process of senescence [71].

4.3 Angiogenesis

Angiogenesis is the formation of new blood vessels from existing vessels. Angiogenesis involves three steps: initiation, sprouting/migration, and maturation. In angiogenesis, the complex signaling pathways between ECs and pericytes which include the platelet-derived growth factor (PDGF)-BB/PDGF receptor beta (PDGFRβ) pathway [72, 73], the angiopoitin1/tyrosine protein kinase receptor (Tie2) signaling pathway [74], the vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR) pathway [46], the sphingosine-1-phosphate (S1P) signaling pathway [75], transforming growth factor beta (TGF-β) [76] are the foundation for the formation and stabilization of new blood vessels. Pericytes regulate the expression of VEGF, resulting in the instability of blood vessels and initiating the angiogenesis [77]. Pericytes detach from blood vessels to pave the way for endothelial sprouting [78]. The migrating ECs secrete VEGF to stabilize nascent vessels and signal to pericytes to recruit VEGF. Moreover, the recruited pericytes communicate with ECs to promote the stabilization and maturation of new blood vessels [78]. The coverage of pericytes is a marker of vascular maturation and a lack of pericytes results in vascular hyperplasia [79].

5. Mechanisms through which Pericytes Regulate Aβ

Aβ is continuously generated by neurons and other cells in the healthy brain, and is subsequently cleared through various pathways [80, 81, 82] including receptor-dependent transport [80, 83, 84, 85, 86], cytosolic protease-mediated intracellular degradation [87] and glymphatic clearance [88]. In AD, the clearance of Aβ is impaired, and an imbalance between Aβ production and clearance leads to the aberrant accumulation of Aβ [7]. Moreover, pericytes play a considerable role in the clearance of Aβ, while the loss of pericytes in AD exacerbates the deposition of Aβ in the parenchyma.

5.1 Pericytes Clear Aβ by LDL Receptor-Related Protein-1 (LRP-1)

LDL receptor-related protein-1 (LRP-1) is an apoE receptor that mediates the clearance of Aβ. Using freshly isolated cortical slices incubated with Aβ, Ma et al. [80] showed that pericytes rapidly remove Cy3-Aβ42. In addition, in AD and APPswe/0 mice, the abundant accumulation of Aβ in pericytes indicates the important role of these cells in clearing Aβ at the BBB. Additionally, in an LRP-1 conditional knockout model, Cy3-Aβ42 uptake by pericytes was reduced by 80% compared with that in the control group. The process of clearing Aβ by pericytes can be inhibited by antibodies against LRP-1 [80, 84], further confirming that the clearance of Aβ is mediated by LRP-1 (Fig. 1A). Compared with that in adult wild-type mice, clearance in apoE knockout mice is substantially reduced, indicating that LRP-1-mediated transport can be influenced by apoE, a definite risk factor for AD [85]. ApoE is required for Aβ clearance and is isoform- specific. A study revealed that apoE3, but not apoE4, normalizes Aβ clearance in mouse pericytes with silenced mouse apoE [80], while another study revealed that the binding of Aβ to apoE3 reduces its clearance rate at the BBB. It has also been reported that Aβ binding to apoJ significantly accelerates the BBB clearance rate [86, 89].

Fig. 1.

The interactions between Aβ and pericytes. (A) LRP-1 and P-gp mediate the efflux of Aβ while RAGE regulate the influx of Aβ into brain parenchyma. BACE1 on pericytes degrades Aβ1-40 into Aβ34 intermediates and the Aβ (1-42)-RAGE interaction induces the generation of VEGF and MCP-1 contributing to the vascular remodeling. (B) CD36 mediates the clearance of Aβ1-40 by pericytes. The reduced expression of CD36 promotes the deposition of Aβ1-40 resulting in CAA. (C) Oligomeric Aβ1-42 activates NOX4 in pericytes to produce ROS and ET in sequence, and ET binds to ETA-R on pericytes, triggering capillary constriction. Capillary constriction results in the reduction of CBF and the glucose and oxygen it contains. Hypoxia in turn upregulates the expression of BACE1, further increasing the generation of Aβ and forming an amplified positive loop, ultimately leading to synapse dysfunction and neuron loss. (D) Fibrillar Aβ1-42 activates MMP-9 to induce NG2 sheds from pericytes leading to the detachment of pericytes and the destruction of endothelial TJs which is the significant part of BBB. (E) Aβ1-40 induces pericytes mitophagy through the CD36/PINK1/Parkin pathway and increases oxidative stress in pericytes. The increased lipid ROS and iron ions caused by oxidative stress in pericytes inducing pericytes ferroptosis dependent on mitochondrial autophagy. (F) Fibrillar Aβ1-40 reduces the viability and proliferation of pericytes, and increases the activity of the key apoptotic proteins caspase3/7 while the effects of monomer Aβ1-40 are completely opposite. LRP-1, LDL receptor-related protein-1; RAGE, receptors for advanced glycation end products; Aβ, beta-amyloid; BACE1, β-site amyloid precursor protein (APP) cleaving enzyme 1; VEGF, vascular endothelial growth factor; CAA, cerebral amyloid angiogenesis; NOX4, nicotinamide adenine dinucleotide phosphate oxidase 4; ROS, reactive oxygen species; ET, endothelin; CBF, cerebral blood flow; MMP-9, matrix metalloproteinase-9; NG2, neural/glial antigen 2; TJs, endothelial tight junctions; BBB, blood–brain barrier; PINK1, PTEN-induced putative kinase 1; sAPPβ, soluble amyloid precursor protein beta; MCP-1, monocyte chemoattractant protein-1; ETA-R, endothelin receptor type A.

The expression of LRP-1 is downregulated in AD patients exacerbating Aβ pathology [90]. By injecting pericytes into APP/PS1 mice, Tachibana et al. [91] showed that microcirculation improved in the pericyte-injected hemisphere and that the deposition of Aβ decreased in a manner dependent on the expression of LRP-1 on pericytes. However, in a recent phase I clinical trial in which mesenchymal stem cells (MSCs) were stereotactically injected to the brains of AD patients, no significant effects on cognitive function were observed. The use of transplanted pericytes or MSCs in the brain to prevent or treat AD has not been validated [92].

5.2 Aβ40 is Degraded into Aβ34 in Pericytes through β-Site Amyloid Precursor Protein (APP) Cleaving Enzyme 1 (BACE1)

Amyloid precursor protein (APP) is cleaved by β-Site Amyloid Precursor Protein (APP) Cleaving Enzyme 1 (BACE1) and γ-secretase, which sequentially results in the formation of Aβ peptides, including Aβ40 and Aβ42 [93]. In addition to “amyloidogenic” activity, BACE1 also possesses “amyloidolytic” activity, whereby it degrades longer Aβ isoforms at position 34 into Aβ34 intermediates [94] (Fig. 1A). A previous study demonstrated that BACE1 is expressed in pericytes [95]. Treating pericytes with a BACE inhibitor resulted in a dose-dependent decrease in Aβ34 levels, indicating the role of BACE1 in the cleavage of Aβ peptides and the formation of Aβ34 in pericytes [95]. Notably, the substrate of BACE1 is Aβ40, but not Aβ42 [94, 95].

In AD, the Aβ34/Aβ40 ratio is decreased significantly, and the level of Aβ34 is correlated with disease progression [96]. The progression of AD can be divided into six stages. AD in Braak stage I-II is clinically silent, AD in Braak stage III–IV is incipient and AD in Braak stage V–VI is fully developed [97, 98, 99]. Kirabali et al. [95] revealed that in Braak stage II, Aβ34 levels peak, while in Braak stages III and IV, the immunoactivity of Aβ34 significantly decreases which explains the dysfunction and loss of brain pericytes in AD pathogenesis. Moreover, analysis of PDGFRβ immunoactivity revealed that the loss of pericytes had already occurred at Braak stage II [37].

5.3 Pericytes Regulate Aβ Clearance via RAGE

AGEs (advanced glycation end-products) are the final products of the nonenzymatic glycation of proteins (Maillard reaction) [100], which is irreversible. AGEs can bind and destroy various histocytes through a process called cross linking. A series of studies have shown that AGEs accelerate aging and cause neurodegenerative disorders including AD [101, 102, 103]. Receptors for advanced glycation end products (RAGE), a multiligand receptor of the immunoglobulin family, can not only specifically bind to AGEs, but also bind to various ligands such as high-mobility group box-1 (HMGB1), S100 and Aβ [104, 105], and play vital roles in the occurrence and development of various diseases. RAGE plays a critical role in regulating the influx of circulating Aβ into the brain via the BBB as a transporter [106]. Moreover, RAGE promotes the generation and accumulation of Aβ by enhancing the activity of β-secretase and γ-secretase [107]. In addition, RAGE induces dysfunction of synapses and neuronal circuits, which is the structural foundation of cognition [108, 109]. According to its structure, RAGE is classified into three isoforms: N-truncated, and C-truncated, which are also called endogenous secretory RAGE (esRAGE) [110]. Moreover, RAGE can be cleaved by proteolytic enzymes to form cRAGE. The soluble form of RAGE (sRAGE) is composed of esRAGE and cRAGE. Notably, sRAGE can interact with Aβ to form sRAGE-Aβ interactions which can inhibit the neurotoxicity of RAGE and promote Aβ clearance from the brain [110].

It has been proven that RAGE is expressed on pericytes [111]. Using small interfering RNA (siRNA) technology to suppress the expression of the pericyte RAGE gene, Lue et al. [111] showed that the level of Aβ dramatically decreased, indicating that the Aβ (1-42)-RAGE interaction may function by tethering Aβ to the cell surface of pericytes, advancing the Aβ-Aβ interaction and further promoting fibrillogenesis. By blocking RAGE with an anti-RAGE antibody, the levels of Aβ-induced VEGF and monocyte chemoattractant protein-1 (MCP-1) were decreased, indicating that RAGE-Aβ interactions in pericytes contribute to the vascular remodeling that is observed in AD (Fig. 1A). The combination of Aβ and pericyte RAGE can induce oxidative stress and a subsequent inflammatory response by activating a variety of signaling pathways, including mitogen-activated protein kinase (MAPK), glycogen synthase kinase 3 (GSK-3) and nuclear factor kappa-B (NF-κB) [112]. Oxidative stress and inflammatory reactions can thicken the BM and increase the deposition of Aβ, eventually leading to vascular amyloidosis and disruption of the BBB [111]. In addition, the Aβ-RAGE interaction results in cognitive impairment by accelerating the aging process and inducing oxidative stress [113]. In AD, elevated RAGE levels may account for neuronal death and cognitive impairment. However, the level of sRAGE is lower [114]. In view of this, RAGE inhibitors may be potential targets for treating AD and these agents have been proven to be effective in preclinical and clinical studies [115], although the results have been unsatisfactory.

5.4 Pericytes Efflux Aβ via P-Glycoprotein (P-gp)

P-gp, a subtype of the ATP binding cassette (ABC) transporter family, is an ATP-dependent transporter responsible for the efflux of various substrates from the brain to the blood [116, 117]. Using an immunogold technique with monoclonal anti-P-gp antibodies, Bendayan et al. [118] showed that gold particles are present in ECs, astrocytes and pericytes, suggesting that pericytes can express P-gp. In human studies, the expression level of P-gp was shown to be negatively correlated with the accumulation of Aβ [119, 120].

Moreover, it has been proven that inhibiting P-gp leads to increased intracellular deposition of Aβ in brain capillaries [121, 122], suggesting that P-gp plays a crucial role in the clearance of Aβ. However, the mechanism by which P-gp affects Aβ transport remains controversial. A variety of studies support the notion that Aβ stimulates the ATPase activity of P-gp [123, 124]. However, Bello et al. [125] reported that Aβ has no effect on the adenosine triphosphate (ATP) hydrolysis activity of P-gp. In previous research, McCormick et al. [123] noted that the activation of P-gp ATPase by Aβ depends on the lipid environment, which may account for the differences between those studies. In addition, Aβ can affect P-gp conversely. By treating transgenic human amyloid precursor protein (hAPP) overexpressing mice with an irreversible inhibitor of the ubiquitin-activating enzyme E1, Hartz et al. [126] showed that retained P-gp results in a decreased level of Aβ, suggesting that Aβ induces P-gp degeneration through the ubiquitination pathway. In addition, the brains of AD patients exhibit marked decrease in P-gp and a significant increase in Aβ deposition and ubiquitinated Aβ [126, 127, 128].

5.5 Pericytes Uptake Aβ1-40 via CD36

CD36, a glycosylated membrane protein, is widely expressed in the nervous system, including in pericytes [129]. CD36 is involved in a variety of pathological processes, such as vascular oxidative stress, the inflammatory response, mitochondrial dysfunction and neurovascular uncoupling [129, 130, 131].

Immunofluorescence staining revealed that, CD36 and Aβ1-40 colocalize with PDGFRβ, a marker of pericytes, suggesting that CD36 may be involved in the clearance of Aβ1-40 by pericytes [132]. In transgenic mice lacking CD36, Li et al. [132] showed a reduction in Aβ1-40 and cerebral amyloid angiopathy (CAA), suggesting that CD36 promotes the deposition of Aβ1-40 resulting in vascular dysfunction (Fig. 1B). Moreover, the transcription and expression levels of CD36 in pericytes treated with Aβ1-40 increased in a concentration-dependent manner aggravating vascular dysfunction [132]. Aβ1-40 increases the permeability of the BBB in vitro, which can be reversed by inhibiting the expression of CD36 in pericytes, suggesting that inhibiting the expression of CD36 increases BBB tightness [133], providing a new therapeutic target for preventing BBB destruction during AD progression.

6. The Effects of Aβ on Pericytes

Aβ exerts toxic effects on pericytes through various pathways, and a significant loss of pericytes has been observed in AD patients. We have summarized the pathological effects of interactions among targets of pericytes and different species of Aβ, as well as the pathological changes observed in AD, in Table 2 (Ref. [36, 37, 128, 134, 135, 136, 137, 138, 139, 140, 141]).

Table 2.Interactions among targets in pericytes and different species of Aβ (f: fibrillar; o: oligomeric; m: monomeric), pathological effects and pathological changes in AD.
Study type Markers of pericytes Targets The species of Aβ Pathological effects Pathological changes in AD Ref.
In vitro PDGFRβ NOX4 oAβ1-42 NOX4 activated by Aβ1-42 induce oxidative stress in pericytes. ROS trigger the generation of ET, which interact with ETA-R on pericytes, triggering strong capillary constriction. The level of ET increases. [36, 134, 135, 136]
In vitro NG2, PDGFRβ and αSMA MMP-9 fAβ1-42 f Aβ1-42 decreases the activity of MMP-9, preventing the detachment of pericytes. In early stage of AD, the level of MMP-9 and sNG2 in CSF are increased. [137, 138, 139]
oAβ1-42 oAβ1-42 increases the activity of MMP-9, promoting the detachment of pericytes.
Both in vitro and vivo PDGFRβ and NG2 CD36/PINK/Parkin Aβ1-40 Aβ1-40 induces ferroptosis of pericytes by activating mitochondrial autophagy. Pericytes exposed to Aβ1-40 exhibit ferroptosis in TEM. [128, 140]
In vitro PDGFRβ and NG2 Caspase3/7 fAβ1-40 f Aβ1-40 reduces the viability and proliferation of pericytes by increasing the activity of caspase 3/7. Aggregated Aβ1-40, the major component of deposition in CAA may account for the loss of pericytes in AD. [37, 141]
mAβ1-40 m Aβ1-40 decreasing the mortality of pericytes by decreasing the activity of caspase 3/7.

Abbreviations: AD, Alzheimer’s disease; PDGFRβ, platelet-derived growth factor receptor-β; NOX4, nicotinamide adenine dinucleotide phosphate oxidase 4; ROS, reactive oxygen species; ET, endothelin; NG2, neural/glial antigen 2; αSMA, alpha-smooth muscle actin; MMP-9, matrix metalloproteinase-9; CSF, cerebrospinal fluid; TEM, transmission electron microscopy; CAA, cerebral amyloid angiogenesis.

6.1 Aβ1-42 Evokes the Constriction of Pericytes

Previous studies have shown that a decrease in the CBF is the earliest change in AD patients [19], and capillaries exhibit focal constriction [142]. Vascular resistance in the brain mainly occurs in capillaries, and the CBF is regulated by pericytes, indicating that pericytes dysfunction contributes to vascular disturbances in AD [22]. One of the most characteristic changes in AD is the aberrant deposition of Aβ, which results in the formation of Aβ plaques in the brain parenchyma, suggesting that Aβ may be the latent culprit.

In human brain slices, Aβ1-42 (oligomeric and monomeric) can trigger a slowly progressive constriction of capillaries near pericytes, suggesting that Aβ1-42 induces the constriction of human pericytes in a concentration-dependent manner within limits [36]. Reactive oxygen species (ROS) are generated by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and can be removed by superoxide dismutase 1 (SOD1) [134]. Recently, Nortley et al. [36] discovered that oligomeric Aβ1-42 evoked capillary constriction could be blocked by endothelin receptor type A (ETA-R), SOD1 and NADPH oxidase inhibitors, suggesting that ROS and endothelin (ET) participate in the constriction induced by Aβ1-42. Moreover, evidence has shown that ROS in pericytes are produced by reduced nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4) [135] rather than other isoforms. After ET was applied along with SOD1, the capillaries remained constricted, demonstrating that ET functions downstream of ROS. Moreover, ET combined with ETA-Rs on pericytes triggers intense constriction [143]. In summary, Aβ1-42 activates NOX4 in pericytes to generate ROS, and these ROS induce the generation of downstream ETs, which then interact with ETA-Rs on pericytes, triggering strong capillary constriction [36, 144]. The constriction of capillaries increases vascular resistance, causing a reduction in the CBF, which leads to a decrease in glucose and oxygen supplies, ultimately leading to synapse dysfunction and neuron loss [145]. In addition, hypoxia in turn upregulates the expression of BACE1, further increasing the generation of Aβ and forming an amplified positive loop [143] (Fig. 1C). The discovery of this mechanism overturns the previous view that the reduction in the CBF is the result of arteriole constriction.

It has been observed that the level of ET increases in AD with the upregulation of enzymes responsible for synthesizing ET [136]. Similarly, compared to those in nondemented controls, Sengillo et al. [69] reported a remarkable loss of pericytes in the cortex and hippocampus of AD patients, which may be attributed to the fact that chronic exposure to Aβ1-42 results in the constriction and rigid death of pericytes. Fortunately, blocking NOX4 and ETA-Rs prevent further contraction caused by Aβ, although the capillary diameter does not return to baseline levels. Moreover, C-type natriuretic peptide (CNP) successfully reverses the Aβ-evoked constriction [136] and may be applied in the treatment of AD in the future.

6.2 Aβ1-42 Induces the Detachment of Pericytes by Activating MMP-9 to Induce NG2 Shedding from Pericytes

Neural/glial antigen 2 (NG2), a transmembrane proteoglycan, is an original marker of pericytes [30]. NG2 not only plays a significant role in the proliferation and motility of pericytes, but also promotes the formation and maturation of endothelial TJs [38].

Aβ1-42 is the principal component of neuritic plaques characterized in AD. Different aggregated forms of Aβ1-42 have been proven to influence the shedding of NG2 from pericytes differently. NG2 sheds pericytes to form soluble NG2 (sNG2). Fibrillar Aβ1-42 decreases the level of sNG2, while the level of sNG2 increases after exposure to oligomer-enriched preparations of Aβ1-42 [137]. After exerting inhibitors of matrix metalloproteinase (MMP, an angiogenic factor secreted by pericytes) [138], the consequences of fibrillar Aβ1-42 remain, while increase in sNG2 resulting from oligomeric Aβ1-42 is eliminated, suggesting that the shedding of NG2 induced by Aβ is mediated by MMP-9. Moreover, fibrillar Aβ1-42 decreases the activity of MMP-9 while oligomeric Aβ1-42 increased MMP-9 activity [137]. However, oligomeric Aβ1-42 does not alter the concentration of MMP-9, indicating that Aβ1-42 affects the activity rather than the secretion of MMP-9.

Taken together, in the early stage of AD, Aβ1-42 exists as oligomers and activates MMP-9, which subsequently increases the level of sNG2. The finding coincides with the discovery of increased MMP-9 in cerebrospinal fluid (CSF) during early AD pathogenesis [139]. sNG2 has been demonstrated to promote angiogenesis, resulting in unstable blood vessels and dysfunction of the BBB [146], and the level of sNG2 is increased in the CSF of AD patients [147]. In addition, the release of NG2 from the cell surface of pericytes drives pericyte detachment and contributes to the loss and dysfunction of TJs. Even worse, this effect could be aggravated by the enhanced degradation of the extracellular matrix and TJs caused by increased MMP-9 activity [148]. The detachment of pericytes and destruction of endothelial TJs destroy the integral structure of the BBB and increase the permeability of the BBB (Fig. 1D). Subsequently, the hazardous substances enter the brain and neurotoxicity caused by Aβ could cause neuronal loss and cognitive decline. As the disease progresses, fibrillar Aβ1-42 accumulates, which may be the protective mechanism of the body [149].

Researchers found that the SIRT1 activator, resveratrol, reduced serum MMP-9 levels in AD patients, thus reducing neuro-inflammation [141]. Additionally, resveratrol was able to slow down the progressive decline in daily living scores (ADLs) in AD patients [141]. In spite of this, the relationship between the delay in cognitive decline and the decrease in MMP-9 remains unknown. The discovery that Aβ1-42 influences the shedding of NG2 on pericytes via MMP-9 may explain the pathology of AD and provide a new therapeutic strategy, such as specifically inhibiting MMP-9, to prevent AD dysfunction.

6.3 Aβ1-40 Induces Pericyte Mitophagy-Dependent Ferroptosis through the CD36/PINK/Parkin Pathway

Aβ1-42 (<10%) and Aβ1-40 (<80%) are two typical soluble monomeric subtypes of Aβ [49]. Aβ1-42 is more likely to form insoluble aggregates than Aβ1-40 [6], but Aβ1-40 can be deposited in the vascular system to form CAA [150]. Recently, it has been reported that more than 90% of patients with AD have CAA and CAA usually precedes the formation of neuritic plaques that are composed mainly of Aβ1-42 [151]. Aβ1-40 is presumed to have a particular toxicity to pericytes [152] and could accelerate the disruption of the BBB.

A recent study showed that pericytes treated with Aβ1-40 exhibit a prominent decrease in proliferation and a marked increas in mitochondrial ROS (Mito SOX) in a manner dependent on both time and concentration [132]. The results also demonstrated that Aβ1-40 activates mitochondrial autophagy through the CD36/PINK/Parkin pathway (PINK, PTEN-induced putative kinase) [132]. Mitochondrial damage and autophagy induced by Aβ1-40 often lead to apoptosis [140]. Notably, Aβ1-40 rather than apoptosis increases oxidative stress in pericytes. An increase in lipid ROS is accompanied by an increase in the concentration of iron ions, indicating that Aβ1-40 may induce pericyte ferroptosis [153]. The morphological features of pericytes exposed to Aβ1-40 according to transmission electron microscopy (TEM) also correspond with ferroptosis. Moreover, inhibiting mitochondrial autophagy prevents pericyte ferroptosis and ferroptosis inhibitors could prevent mitochondrial autophagy evoked by Aβ1-40, suggesting that ferroptosis is dependent on mitochondrial-related autophagy [132] (Fig. 1E).

6.4 Aβ1-40 Affects Pericytes in an Aggregation-Dependent Manner

The expression of pericyte markers is dynamic, and depends on the functional state of pericytes; for example, PDGFRβ labels relatively immature pericytes [33], NG2 stimulates the proliferation and migration of pericytes [34], and laminin is expressed in active or mature subsets of pericytes [154]. In the hippocampus of AD patients, a significant reduction in the number of pericytes expressing NG2 [37], CD13 and PDGFRβ [70] is observed, while the number of pericytes expressing other markers is not affected. It can be concluded that the subsets of pericytes that participate in activation, migration, and proliferation are affected by the pathology of AD. A previous study demonstrated that the CAA formed by Aβ1-40 is associated with pericyte degenerative changes [150]. Interestingly, Aβ1-40 levels are associated with the number of pericytes [37]. Since aggregated Aβ1-40 is toxic [150], it is speculated that monomeric Aβ1-40 may be beneficial.

Schultz et al. [37] showed that fibrillar Aβ1-40 reduces the viability and proliferation of pericytes in vitro, and increases the activity of the key apoptotic proteins caspase3/7. These findings are consistent with the discovery of pericyte degeneration near fibrillar Aβ1-40 [150]. Moreover, the monomer Aβ1-40 decreases the mortality of pericytes by decreasing the activity of caspase3/7, and promoting pericyte proliferation, which could explain the positive correlation between the levels of monomeric Aβ1-40 and the number of pericytes (Fig. 1F).

The occurrence of CAA and AD largely overlap [13, 14, 15] and the occurrence of advanced CAA is related to more severe cognitive impairment in patients with AD [15, 16]. Aggregated Aβ1-40, the major component of deposition in the CAA may account for the loss of pericytes in AD patients [155].

7. Strategies to Prevent or Treat AD via Pericytes

Further understanding of the interactions between pericytes and Aβ could lead to new insights for the treatment of AD. Superoxide dismutase-1 (SOD1) can eliminate ROS generated by NOX4 activated by Aβ [36]. Using SOD1 to eliminate ROS may be effective at preventing the constriction of pericytes. Indeed, overexpression of SOD1 or topical application of exogenous SOD could reverse vascular dysfunction and premature mortality in transgenic mice overexpressing APP [156]. Pterostilbene (PTE), the natural dimethylated analog of resveratrol, can upregulate the expression of SIRT1 and SOD to exert neuroprotective effects [157]. Moreover, since Aβ cannot induce the constriction of pericytes without ET [143], reducing the generation of ET or blocking the combination of ET and ETA-R may be effective. It has been proven that blocking NOX4 or ETA-Rs could prevent further constriction of capillaries evoked by Aβ and CNPs could successfully reverse the constriction of capillaries mediated by ET [36]. In addition, Aβ induces endothelial dysfunction characterized by attenuated endothelium-dependent relaxation and increased endothelium-dependent constriction since Aβ inactivates vasodilators produced by the endothelium and increases the production and release of ET [158]. In APP overexpressing mice, bosentan, an antagonist of both ETA and the ETB receptor, was shown to preserve the endothelial function of the aorta and carotids [159]. Autopsy evidence has shown that the majority of Alzheimer’s disease patients suffer from vascular diseases such as CAA [13, 15]. Several ET receptor antagonists, including bosentan have been applied to treat pulmonary hypertension, and recent studies have demonstrated the beneficial effects of bosentan in restoring the cerebrovascular function of diabetic rats and preventing coronary endothelial functions in hypercholesterolemic pigs [160, 161, 162]. The ability of bosentan to preserve endothelial functions in Aβ overexpressing Tg2576 mice demonstrated the potential of ET receptor antagonists for the prevention and treatment of AD.

Another promising approach might be to implant pericytes or mesenchymal stem cells (MSCs) into the brain. In APP/PS1 mice, the CBF was increased and Aβ plaques were significantly reduced in the pericyte-injected hemisphere [91]. However, no significant effects on cognitive function were observed in a recent phase I clinical trial in which stereotactically injected MSCs were administered to the brains of AD patients [92]. Many studies have shown that RAGE inhibitors may be potential targets for treating AD [107, 163, 164]. Recently, PF-04494700, an oral inhibitor of RAGE has attracted widespread attention. However, two clinical trials on PF-04494700 have shown that although PF-04494700 is safe and well-tolerated, it has no apparent benefit in improving cognitive decline [165, 166]. Notably, high-dose PF-04494700 could also increase adverse reactions and exacerbate cognitive impairment [165].

8. Conclusion

Pericytes are multifunctional cells of the vascular system and important components of the BBB and NVU. Pericytes regulate the CBF in response to neurotransmitters and neuronal activity and are essential for endothelial TJs and they are necessary for the formation and maintenance of the BBB. Bidirectional communication between ECs and pericytes is necessary for angiogenesis. Moreover, pericytes mediate phagocytosis to maintain homeostasis in the brain.

The aberrant deposition of Aβ is the predominant pathological change in AD. Pericytes can clear Aβ via LRP-1, RAGE, P-gp, and CD36, and Aβ is degraded in pericytes via BACE1. The impairment of these pathways may account for the pathogenesis of AD. Aβ1-42 evokes the constriction of pericytes and causes death after chronic exposure. A decrease in the CBF caused by capillary constriction leads to hypoxia and glucose deficiency, contributing to the neuronal dysfunction and cognitive decline. Aβ1-42 also induces the detachment of pericytes. Aβ1-40 induces pericyte mitophagy-dependent ferroptosis through the CD36/PINK/Parkin pathway. The loss of pericytes exacerbates the aggregation of Aβ in AD. Conversely, the loss of pericytes is a result of the action of Aβ. Further exploration of the interactions between pericytes and Aβ is beneficial for understanding AD and provides new therapeutic targets for the prevention and treatment of AD.

Author Contributions

YYL—literature collection, literature analysis and drafting the manuscript; DDG—literature collection and literature analysis; RND—editing, preparing the figures, reviewing and giving final approval of the version; YL—editing, preparing the figures, reviewing and giving final approval of the version. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This work is supported by National Natural Science Foundation of China (No. 82101487), Natural Science Foundation of Shandong Province, China (ZR2021QH161) and Taishan Scholar Program of Shandong Province (tsqn202211318).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Scheltens P, De Strooper B, Kivipelto M, Holstege H, Chételat G, Teunissen CE, et al. Alzheimer’s disease. Lancet. 2021; 397: 1577–1590.
[2]
Jack CR, Jr, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimer’s & Dementia. 2018; 14: 535–562.
[3]
Chen X, Firulyova M, Manis M, Herz J, Smirnov I, Aladyeva E, et al. Microglia-mediated T cell infiltration drives neurodegeneration in tauopathy. Nature. 2023; 615: 668–677.
[4]
Greenberg SM, Bacskai BJ, Hernandez-Guillamon M, Pruzin J, Sperling R, van Veluw SJ. Cerebral amyloid angiopathy and Alzheimer disease - one peptide, two pathways. Nature Reviews. Neurology. 2020; 16: 30–42.
[5]
Inoue Y, Ando Y, Misumi Y, Ueda M. Current Management and Therapeutic Strategies for Cerebral Amyloid Angiopathy. International Journal of Molecular Sciences. 2021; 22: 3869.
[6]
Dawkins E, Small DH. Insights into the physiological function of the β-amyloid precursor protein: beyond Alzheimer’s disease. Journal of Neurochemistry. 2014; 129: 756–769.
[7]
Tarasoff-Conway JM, Carare RO, Osorio RS, Glodzik L, Butler T, Fieremans E, et al. Clearance systems in the brain-implications for Alzheimer disease. Nature Reviews. Neurology. 2015; 11: 457–470.
[8]
Gkanatsiou E, Portelius E, Toomey CE, Blennow K, Zetterberg H, Lashley T, et al. A distinct brain beta amyloid signature in cerebral amyloid angiopathy compared to Alzheimer’s disease. Neuroscience Letters. 2019; 701: 125–131.
[9]
Hawkes CA, Härtig W, Kacza J, Schliebs R, Weller RO, Nicoll JA, et al. Perivascular drainage of solutes is impaired in the ageing mouse brain and in the presence of cerebral amyloid angiopathy. Acta Neuropathologica. 2011; 121: 431–443.
[10]
Arbel-Ornath M, Hudry E, Eikermann-Haerter K, Hou S, Gregory JL, Zhao L, et al. Interstitial fluid drainage is impaired in ischemic stroke and Alzheimer’s disease mouse models. Acta Neuropathologica. 2013; 126: 353–364.
[11]
Martin L, Bouvet P, Chounlamountri N, Watrin C, Besançon R, Pinatel D, et al. VEGF counteracts amyloid-β-induced synaptic dysfunction. Cell Reports. 2021; 35: 109121.
[12]
Peng L, Bestard-Lorigados I, Song W. The synapse as a treatment avenue for Alzheimer’s Disease. Molecular Psychiatry. 2022; 27: 2940–2949.
[13]
Boyle PA, Yu L, Wilson RS, Leurgans SE, Schneider JA, Bennett DA. Person-specific contribution of neuropathologies to cognitive loss in old age. Annals of Neurology. 2018; 83: 74–83.
[14]
Arvanitakis Z, Leurgans SE, Wang Z, Wilson RS, Bennett DA, Schneider JA. Cerebral amyloid angiopathy pathology and cognitive domains in older persons. Annals of Neurology. 2011; 69: 320–327.
[15]
Brenowitz WD, Nelson PT, Besser LM, Heller KB, Kukull WA. Cerebral amyloid angiopathy and its co-occurrence with Alzheimer’s disease and other cerebrovascular neuropathologic changes. Neurobiology of Aging. 2015; 36: 2702–2708.
[16]
Goos JDC, Kester MI, Barkhof F, Klein M, Blankenstein MA, Scheltens P, et al. Patients with Alzheimer disease with multiple microbleeds: relation with cerebrospinal fluid biomarkers and cognition. Stroke. 2009; 40: 3455–3460.
[17]
Jäger HR. The connection between cerebral amyloid angiopathy and Alzheimer’s disease. European Radiology. 2023. (online ahead of print)
[18]
Kim SH, Ahn JH, Yang H, Lee P, Koh GY, Jeong Y. Cerebral amyloid angiopathy aggravates perivascular clearance impairment in an Alzheimer’s disease mouse model. Acta Neuropathologica Communications. 2020; 8: 181.
[19]
Iturria-Medina Y, Sotero RC, Toussaint PJ, Mateos-Pérez JM, Evans AC, Alzheimer’s Disease Neuroimaging Initiative. Early role of vascular dysregulation on late-onset Alzheimer’s disease based on multifactorial data-driven analysis. Nature Communications. 2016; 7: 11934.
[20]
Agnollitto MISS, Leoni RF, Foss MP, Palaretti J, Cayres M, Pansarim V, et al. Influence of cerebral blood flow on volumetric loss related to Alzheimer’s disease. Dementia & Neuropsychologia. 2023; 17: e20230004.
[21]
Qiu M, Zhou D, Zhu H, Shao Y, Li Y, Wang Y, et al. Alterations of Cerebral Blood Flow and its Connectivity Patterns Measured with Arterial Spin Labeling in Mild Cognitive Impairment. Current Alzheimer Research. 2023; 20: 567–576.
[22]
Gould IG, Tsai P, Kleinfeld D, Linninger A. The capillary bed offers the largest hemodynamic resistance to the cortical blood supply. Journal of Cerebral Blood Flow and Metabolism. 2017; 37: 52–68.
[23]
Mangialasche F, Solomon A, Winblad B, Mecocci P, Kivipelto M. Alzheimer’s disease: clinical trials and drug development. The Lancet. Neurology. 2010; 9: 702–716.
[24]
Finney AC, Orr AW. Guidance Molecules in Vascular Smooth Muscle. Frontiers in Physiology. 2018; 9: 1311.
[25]
Grant RI, Hartmann DA, Underly RG, Berthiaume AA, Bhat NR, Shih AY. Organizational hierarchy and structural diversity of microvascular pericytes in adult mouse cortex. Journal of Cerebral Blood Flow and Metabolism. 2019; 39: 411–425.
[26]
Sweeney MD, Ayyadurai S, Zlokovic BV. Pericytes of the neurovascular unit: key functions and signaling pathways. Nature Neuroscience. 2016; 19: 771–783.
[27]
Attwell D, Mishra A, Hall CN, O’Farrell FM, Dalkara T. What is a pericyte? Journal of Cerebral Blood Flow and Metabolism. 2016; 36: 451–455.
[28]
Bondjers C, He L, Takemoto M, Norlin J, Asker N, Hellström M, et al. Microarray analysis of blood microvessels from PDGF-B and PDGF-Rbeta mutant mice identifies novel markers for brain pericytes. FASEB Journal. 2006; 20: 1703–1705.
[29]
Hartmann DA, Underly RG, Grant RI, Watson AN, Lindner V, Shih AY. Pericyte structure and distribution in the cerebral cortex revealed by high-resolution imaging of transgenic mice. Neurophotonics. 2015; 2: 041402.
[30]
He L, Vanlandewijck M, Raschperger E, Andaloussi Mäe M, Jung B, Lebouvier T, et al. Analysis of the brain mural cell transcriptome. Scientific Reports. 2016; 6: 35108.
[31]
Sá da Bandeira D, Kilpatrick AM, Marques M, Gomez-Salazar M, Ventura T, Gonzalez ZN, et al. PDGFRβ+ cells play a dual role as hematopoietic precursors and niche cells during mouse ontogeny. Cell Reports. 2022; 40: 111114.
[32]
Smyth LCD, Rustenhoven J, Scotter EL, Schweder P, Faull RLM, Park TIH, et al. Markers for human brain pericytes and smooth muscle cells. Journal of Chemical Neuroanatomy. 2018; 92: 48–60.
[33]
Göritz C, Dias DO, Tomilin N, Barbacid M, Shupliakov O, Frisén J. A pericyte origin of spinal cord scar tissue. Science. 2011; 333: 238–242.
[34]
Jung B, Arnold TD, Raschperger E, Gaengel K, Betsholtz C. Visualization of vascular mural cells in developing brain using genetically labeled transgenic reporter mice. Journal of Cerebral Blood Flow and Metabolism. 2018; 38: 456–468.
[35]
Armulik A, Genové G, Betsholtz C. Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Developmental Cell. 2011; 21: 193–215.
[36]
Nortley R, Korte N, Izquierdo P, Hirunpattarasilp C, Mishra A, Jaunmuktane Z, et al. Amyloid β oligomers constrict human capillaries in Alzheimer’s disease via signaling to pericytes. Science. 2019; 365: eaav9518.
[37]
Schultz N, Brännström K, Byman E, Moussaud S, Nielsen HM, Netherlands Brain Bank, et al. Amyloid-beta 1-40 is associated with alterations in NG2+ pericyte population ex vivo and in vitro. Aging Cell. 2018; 17: e12728.
[38]
Winkler EA, Bell RD, Zlokovic BV. Central nervous system pericytes in health and disease. Nature Neuroscience. 2011; 14: 1398–1405.
[39]
Zlokovic BV. The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron. 2008; 57: 178–201.
[40]
Zlokovic BV. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nature Reviews. Neuroscience. 2011; 12: 723–738.
[41]
Kisler K, Nelson AR, Montagne A, Zlokovic BV. Cerebral blood flow regulation and neurovascular dysfunction in Alzheimer disease. Nature Reviews. Neuroscience. 2017; 18: 419–434.
[42]
Winkler EA, Bell RD, Zlokovic BV. Pericyte-specific expression of PDGF beta receptor in mouse models with normal and deficient PDGF beta receptor signaling. Molecular Neurodegeneration. 2010; 5: 32.
[43]
Maddaluno L, Rudini N, Cuttano R, Bravi L, Giampietro C, Corada M, et al. EndMT contributes to the onset and progression of cerebral cavernous malformations. Nature. 2013; 498: 492–496.
[44]
Alex L, Tuleta I, Hernandez SC, Hanna A, Venugopal H, Astorkia M, et al. Cardiac Pericytes Acquire a Fibrogenic Phenotype and Contribute to Vascular Maturation After Myocardial Infarction. Circulation. 2023; 148: 882–898.
[45]
Uemura A, Ogawa M, Hirashima M, Fujiwara T, Koyama S, Takagi H, et al. Recombinant angiopoietin-1 restores higher-order architecture of growing blood vessels in mice in the absence of mural cells. The Journal of Clinical Investigation. 2002; 110: 1619–1628.
[46]
Franco M, Roswall P, Cortez E, Hanahan D, Pietras K. Pericytes promote endothelial cell survival through induction of autocrine VEGF-A signaling and Bcl-w expression. Blood. 2011; 118: 2906–2917.
[47]
Hultman K, Strickland S, Norris EH. The APOE ɛ4/ɛ4 genotype potentiates vascular fibrin(ogen) deposition in amyloid-laden vessels in the brains of Alzheimer’s disease patients. Journal of Cerebral Blood Flow and Metabolism. 2013; 33: 1251–1258.
[48]
Halliday MR, Rege SV, Ma Q, Zhao Z, Miller CA, Winkler EA, et al. Accelerated pericyte degeneration and blood-brain barrier breakdown in apolipoprotein E4 carriers with Alzheimer’s disease. Journal of Cerebral Blood Flow and Metabolism. 2016; 36: 216–227.
[49]
Armulik A, Genové G, Mäe M, Nisancioglu MH, Wallgard E, Niaudet C, et al. Pericytes regulate the blood-brain barrier. Nature. 2010; 468: 557–561.
[50]
Zheng Z, Chopp M, Chen J. Multifaceted roles of pericytes in central nervous system homeostasis and disease. Journal of Cerebral Blood Flow and Metabolism. 2020; 40: 1381–1401.
[51]
Soto-Rojas LO, Pacheco-Herrero M, Martínez-Gómez PA, Campa-Córdoba BB, Apátiga-Pérez R, Villegas-Rojas MM, et al. The Neurovascular Unit Dysfunction in Alzheimer’s Disease. International Journal of Molecular Sciences. 2021; 22: 2022.
[52]
Hall CN, Reynell C, Gesslein B, Hamilton NB, Mishra A, Sutherland BA, et al. Capillary pericytes regulate cerebral blood flow in health and disease. Nature. 2014; 508: 55–60.
[53]
Iadecola C. The Neurovascular Unit Coming of Age: A Journey through Neurovascular Coupling in Health and Disease. Neuron. 2017; 96: 17–42.
[54]
Peppiatt CM, Howarth C, Mobbs P, Attwell D. Bidirectional control of CNS capillary diameter by pericytes. Nature. 2006; 443: 700–704.
[55]
Korte N, James G, You H, Hirunpattarasilp C, Christie I, Sethi H, et al. Noradrenaline released from locus coeruleus axons contracts cerebral capillary pericytes via α2 adrenergic receptors. Journal of Cerebral Blood Flow and Metabolism. 2023; 43: 1142–1152.
[56]
Lee YJ, Jeon HY, Lee AJ, Kim M, Ha KS. Dopamine ameliorates hyperglycemic memory-induced microvascular dysfunction in diabetic retinopathy. FASEB Journal. 2022; 36: e22643.
[57]
Wu K, Cheng T, Zhai Z, Jiang C, Zhou X. Alpha 7-nicotinic cholinoceptor regulation of pericyte-containing retinal capillaries. British Journal of Pharmacology. 2023; 180: 2196–2213.
[58]
Sun J, Huang Y, Gong J, Wang J, Fan Y, Cai J, et al. Transplantation of hPSC-derived pericyte-like cells promotes functional recovery in ischemic stroke mice. Nature Communications. 2020; 11: 5196.
[59]
Yemisci M, Gursoy-Ozdemir Y, Vural A, Can A, Topalkara K, Dalkara T. Pericyte contraction induced by oxidative-nitrative stress impairs capillary reflow despite successful opening of an occluded cerebral artery. Nature Medicine. 2009; 15: 1031–1037.
[60]
Li P, Fan H. Pericyte Loss in Diseases. Cells. 2023; 12: 1931.
[61]
Lin S, Landon B, Zhang H, Jin K. Pericyte Dysfunction Contributes to Vascular Cognitive Impairment Induced by Chronic Cerebral Hypoperfusion in Rats. Aging and Disease. 2023. (online ahead of print)
[62]
Sagare AP, Bell RD, Zhao Z, Ma Q, Winkler EA, Ramanathan A, et al. Pericyte loss influences Alzheimer-like neurodegeneration in mice. Nature Communications. 2013; 4: 2932.
[63]
Abbott NJ, Patabendige AAK, Dolman DEM, Yusof SR, Begley DJ. Structure and function of the blood-brain barrier. Neurobiology of Disease. 2010; 37: 13–25.
[64]
Wu D, Chen Q, Chen X, Han F, Chen Z, Wang Y. The blood-brain barrier: structure, regulation, and drug delivery. Signal Transduction and Targeted Therapy. 2023; 8: 217.
[65]
Zhang ZS, Zhou HN, He SS, Xue MY, Li T, Liu LM. Research advances in pericyte function and their roles in diseases. Chinese Journal of Traumatology. 2020; 23: 89–95.
[66]
Stewart PA, Wiley MJ. Developing nervous tissue induces formation of blood-brain barrier characteristics in invading endothelial cells: a study using quail–chick transplantation chimeras. Developmental Biology. 1981; 84: 183–192.
[67]
Daneman R, Zhou L, Kebede AA, Barres BA. Pericytes are required for blood-brain barrier integrity during embryogenesis. Nature. 2010; 468: 562–566.
[68]
Alkhalifa AE, Al-Ghraiybah NF, Odum J, Shunnarah JG, Austin N, Kaddoumi A. Blood-Brain Barrier Breakdown in Alzheimer’s Disease: Mechanisms and Targeted Strategies. International Journal of Molecular Sciences. 2023; 24: 16288.
[69]
Sengillo JD, Winkler EA, Walker CT, Sullivan JS, Johnson M, Zlokovic BV. Deficiency in mural vascular cells coincides with blood-brain barrier disruption in Alzheimer’s disease. Brain Pathology. 2013; 23: 303–310.
[70]
Li Y, Lu J, Hou Y, Huang S, Pei G. Alzheimer’s Amyloid-β Accelerates Human Neuronal Cell Senescence Which Could Be Rescued by Sirtuin-1 and Aspirin. Frontiers in Cellular Neuroscience. 2022; 16: 906270.
[71]
Stamatovic SM, Martinez-Revollar G, Hu A, Choi J, Keep RF, Andjelkovic AV. Decline in Sirtuin-1 expression and activity plays a critical role in blood-brain barrier permeability in aging. Neurobiology of Disease. 2019; 126: 105–116.
[72]
Adams RH, Alitalo K. Molecular regulation of angiogenesis and lymphangiogenesis. Nature Reviews. Molecular Cell Biology. 2007; 8: 464–478.
[73]
Kemp SS, Lin PK, Sun Z, Castaño MA, Yrigoin K, Penn MR, et al. Molecular basis for pericyte-induced capillary tube network assembly and maturation. Frontiers in Cell and Developmental Biology. 2022; 10: 943533.
[74]
Teichert M, Milde L, Holm A, Stanicek L, Gengenbacher N, Savant S, et al. Pericyte-expressed Tie2 controls angiogenesis and vessel maturation. Nature Communications. 2017; 8: 16106.
[75]
Rosen H, Stevens RC, Hanson M, Roberts E, Oldstone MBA. Sphingosine-1-phosphate and its receptors: structure, signaling, and influence. Annual Review of Biochemistry. 2013; 82: 637–662.
[76]
Armulik A, Abramsson A, Betsholtz C. Endothelial/pericyte interactions. Circulation Research. 2005; 97: 512–523.
[77]
Wu Y, Fu J, Huang Y, Duan R, Zhang W, Wang C, et al. Biology and function of pericytes in the vascular microcirculation. Animal Models and Experimental Medicine. 2023; 6: 337–345.
[78]
Bergers G, Song S. The role of pericytes in blood-vessel formation and maintenance. Neuro-oncology. 2005; 7: 452–464.
[79]
Cuervo H, Mühleder S, García-Gónzalez I, Benedito R. Notch-mediated cellular interactions between vascular cells. Current Opinion in Cell Biology. 2023; 85: 102254.
[80]
Ma Q, Zhao Z, Sagare AP, Wu Y, Wang M, Owens NC, et al. Blood-brain barrier-associated pericytes internalize and clear aggregated amyloid-β42 by LRP1-dependent apolipoprotein E isoform-specific mechanism. Molecular Neurodegeneration. 2018; 13: 57.
[81]
Bateman RJ, Munsell LY, Morris JC, Swarm R, Yarasheski KE, Holtzman DM. Human amyloid-beta synthesis and clearance rates as measured in cerebrospinal fluid in vivo. Nature Medicine. 2006; 12: 856–861.
[82]
Chachaj A, Gąsiorowski K, Szuba A, Sieradzki A, Leszek J. The Lymphatic System In The Brain Clearance Mechanisms - New Therapeutic Perspectives For Alzheimer’s Disease. Current Neuropharmacology. 2023; 21: 380–391.
[83]
Shibata M, Yamada S, Kumar SR, Calero M, Bading J, Frangione B, et al. Clearance of Alzheimer’s amyloid-ss(1-40) peptide from brain by LDL receptor-related protein-1 at the blood-brain barrier. The Journal of Clinical Investigation. 2000; 106: 1489–1499.
[84]
Bell RD, Deane R, Chow N, Long X, Sagare A, Singh I, et al. SRF and myocardin regulate LRP-mediated amyloid-beta clearance in brain vascular cells. Nature Cell Biology. 2009; 11: 143–153.
[85]
Verghese PB, Castellano JM, Holtzman DM. Apolipoprotein E in Alzheimer’s disease and other neurological disorders. The Lancet. Neurology. 2011; 10: 241–252.
[86]
Bell RD, Sagare AP, Friedman AE, Bedi GS, Holtzman DM, Deane R, et al. Transport pathways for clearance of human Alzheimer’s amyloid beta-peptide and apolipoproteins E and J in the mouse central nervous system. Journal of Cerebral Blood Flow and Metabolism. 2007; 27: 909–918.
[87]
Iwata N, Tsubuki S, Takaki Y, Watanabe K, Sekiguchi M, Hosoki E, et al. Identification of the major Abeta1-42-degrading catabolic pathway in brain parenchyma: suppression leads to biochemical and pathological deposition. Nature Medicine. 2000; 6: 143–150.
[88]
Yin M, Pu T, Wang L, Marshall C, Wu T, Xiao M. Astroglial water channel aquaporin 4-mediated glymphatic clearance function: A determined factor for time-sensitive treatment of aerobic exercise in patients with Alzheimer’s disease. Medical Hypotheses. 2018; 119: 18–21.
[89]
Zandl-Lang M, Fanaee-Danesh E, Sun Y, Albrecher NM, Gali CC, Čančar I, et al. Regulatory effects of simvastatin and apoJ on APP processing and amyloid-β clearance in blood-brain barrier endothelial cells. Biochimica et Biophysica Acta. Molecular and Cell Biology of Lipids. 2018; 1863: 40–60.
[90]
Cai Z, Qiao PF, Wan CQ, Cai M, Zhou NK, Li Q. Role of Blood-Brain Barrier in Alzheimer’s Disease. Journal of Alzheimer’s Disease. 2018; 63: 1223–1234.
[91]
Tachibana M, Yamazaki Y, Liu CC, Bu G, Kanekiyo T. Pericyte implantation in the brain enhances cerebral blood flow and reduces amyloid-β pathology in amyloid model mice. Experimental Neurology. 2018; 300: 13–21.
[92]
Kim HJ, Seo SW, Chang JW, Lee JI, Kim CH, Chin J, et al. Stereotactic brain injection of human umbilical cord blood mesenchymal stem cells in patients with Alzheimer’s disease dementia: A phase 1 clinical trial. Alzheimer’s & Dementia. 2015; 1: 95–102.
[93]
Olsson F, Schmidt S, Althoff V, Munter LM, Jin S, Rosqvist S, et al. Characterization of intermediate steps in amyloid beta (Aβ) production under near-native conditions. The Journal of Biological Chemistry. 2014; 289: 1540–1550.
[94]
Ulku I, Liebsch F, Akerman SC, Schulz JF, Kulic L, Hock C, et al. Mechanisms of amyloid-β34 generation indicate a pivotal role for BACE1 in amyloid homeostasis. Scientific Reports. 2023; 13: 2216.
[95]
Kirabali T, Rigotti S, Siccoli A, Liebsch F, Shobo A, Hock C, et al. The amyloid-β degradation intermediate Aβ34 is pericyte-associated and reduced in brain capillaries of patients with Alzheimer’s disease. Acta Neuropathologica Communications. 2019; 7: 194.
[96]
Liebsch F, Kulic L, Teunissen C, Shobo A, Ulku I, Engelschalt V, et al. Aβ34 is a BACE1-derived degradation intermediate associated with amyloid clearance and Alzheimer’s disease progression. Nature Communications. 2019; 10: 2240.
[97]
Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathologica. 1991; 82: 239–259.
[98]
Braak H, Braak E. Staging of Alzheimer’s disease-related neurofibrillary changes. Neurobiology of Aging. 1995; 16: 271–278; discussion 278–284.
[99]
Trejo-Lopez JA, Yachnis AT, Prokop S. Neuropathology of Alzheimer’s Disease. Neurotherapeutics. 2022; 19: 173–185.
[100]
Muthyalaiah YS, Jonnalagadda B, John CM, Arockiasamy S. Impact of Advanced Glycation End products (AGEs) and its receptor (RAGE) on cancer metabolic signaling pathways and its progression. Glycoconjugate Journal. 2021; 38: 717–734.
[101]
D’Cunha NM, Sergi D, Lane MM, Naumovski N, Gamage E, Rajendran A, et al. The Effects of Dietary Advanced Glycation End-Products on Neurocognitive and Mental Disorders. Nutrients. 2022; 14: 2421.
[102]
Moldogazieva NT, Mokhosoev IM, Mel’nikova TI, Porozov YB, Terentiev AA. Oxidative Stress and Advanced Lipoxidation and Glycation End Products (ALEs and AGEs) in Aging and Age-Related Diseases. Oxidative Medicine and Cellular Longevity. 2019; 2019: 3085756.
[103]
Luevano-Contreras C, Chapman-Novakofski K. Dietary advanced glycation end products and aging. Nutrients. 2010; 2: 1247–1265.
[104]
Wautier MP, Tessier FJ, Wautier JL. Advanced glycation end products: A risk factor for human health. Annales Pharmaceutiques Françaises. 2014; 72: 400–408. (In French)
[105]
Paudel YN, Angelopoulou E, Piperi C, Othman I, Aamir K, Shaikh MF. Impact of HMGB1, RAGE, and TLR4 in Alzheimer’s Disease (AD): From Risk Factors to Therapeutic Targeting. Cells. 2020; 9: 383.
[106]
Sharma HS, Castellani RJ, Smith MA, Sharma A. The blood-brain barrier in Alzheimer’s disease: novel therapeutic targets and nanodrug delivery. International Review of Neurobiology. 2012; 102: 47–90.
[107]
Cai Z, Liu N, Wang C, Qin B, Zhou Y, Xiao M, et al. Role of RAGE in Alzheimer’s Disease. Cellular and Molecular Neurobiology. 2016; 36: 483–495.
[108]
Wang YY, Zhou YN, Jiang L, Wang S, Zhu L, Zhang SS, et al. Long-term voluntary exercise inhibited AGE/RAGE and microglial activation and reduced the loss of dendritic spines in the hippocampi of APP/PS1 transgenic mice. Experimental Neurology. 2023; 363: 114371.
[109]
Flores-Muñoz C, Gómez B, Mery E, Mujica P, Gajardo I, Córdova C, et al. Acute Pannexin 1 Blockade Mitigates Early Synaptic Plasticity Defects in a Mouse Model of Alzheimer’s Disease. Frontiers in Cellular Neuroscience. 2020; 14: 46.
[110]
Grossin N, Boulanger E, Wautier MP, Wautier JL. The different isoforms of the receptor for advanced glycation end products are modulated by pharmacological agents. Clinical Hemorheology and Microcirculation. 2010; 45: 143–153.
[111]
Lue LF, Yan SD, Stern DM, Walker DG. Preventing activation of receptor for advanced glycation endproducts in Alzheimer’s disease. Current Drug Targets. CNS and Neurological Disorders. 2005; 4: 249–266.
[112]
Guan L, Mao Z, Yang S, Wu G, Chen Y, Yin L, et al. Dioscin alleviates Alzheimer’s disease through regulating RAGE/NOX4 mediated oxidative stress and inflammation. Biomedicine & Pharmacotherapy. 2022; 152: 113248.
[113]
Yan SD, Zhu H, Zhu A, Golabek A, Du H, Roher A, et al. Receptor-dependent cell stress and amyloid accumulation in systemic amyloidosis. Nature Medicine. 2000; 6: 643–651.
[114]
Daborg J, von Otter M, Sjölander A, Nilsson S, Minthon L, Gustafson DR, et al. Association of the RAGE G82S polymorphism with Alzheimer’s disease. Journal of Neural Transmission. 2010; 117: 861–867.
[115]
Shen CY, Lu CH, Wu CH, Li KJ, Kuo YM, Hsieh SC, et al. The Development of Maillard Reaction, and Advanced Glycation End Product (AGE)-Receptor for AGE (RAGE) Signaling Inhibitors as Novel Therapeutic Strategies for Patients with AGE-Related Diseases. Molecules. 2020; 25: 5591.
[116]
Ambudkar SV, Kimchi-Sarfaty C, Sauna ZE, Gottesman MM. P-glycoprotein: from genomics to mechanism. Oncogene. 2003; 22: 7468–7485.
[117]
Storck SE, Hartz AMS, Bernard J, Wolf A, Kachlmeier A, Mahringer A, et al. The concerted amyloid-beta clearance of LRP1 and ABCB1/P-gp across the blood-brain barrier is linked by PICALM. Brain, Behavior, and Immunity. 2018; 73: 21–33.
[118]
Bendayan R, Ronaldson PT, Gingras D, Bendayan M. In situ localization of P-glycoprotein (ABCB1) in human and rat brain. The Journal of Histochemistry and Cytochemistry. 2006; 54: 1159–1167.
[119]
Chai AB, Leung GKF, Callaghan R, Gelissen IC. P-glycoprotein: a role in the export of amyloid-β in Alzheimer’s disease? The FEBS Journal. 2020; 287: 612–625.
[120]
Vogelgesang S, Cascorbi I, Schroeder E, Pahnke J, Kroemer HK, Siegmund W, et al. Deposition of Alzheimer’s beta-amyloid is inversely correlated with P-glycoprotein expression in the brains of elderly non-demented humans. Pharmacogenetics. 2002; 12: 535–541.
[121]
Tai LM, Loughlin AJ, Male DK, Romero IA. P-glycoprotein and breast cancer resistance protein restrict apical-to-basolateral permeability of human brain endothelium to amyloid-beta. Journal of Cerebral Blood Flow and Metabolism. 2009; 29: 1079–1083.
[122]
Fox E, Bates SE. Tariquidar (XR9576): a P-glycoprotein drug efflux pump inhibitor. Expert Review of Anticancer Therapy. 2007; 7: 447–459.
[123]
McCormick JW, Ammerman L, Chen G, Vogel PD, Wise JG. Transport of Alzheimer’s associated amyloid-β catalyzed by P-glycoprotein. PLoS ONE. 2021; 16: e0250371.
[124]
Lam FC, Liu R, Lu P, Shapiro AB, Renoir JM, Sharom FJ, et al. beta-Amyloid efflux mediated by p-glycoprotein. Journal of Neurochemistry. 2001; 76: 1121–1128.
[125]
Bello I, Salerno M. Evidence against a role of P-glycoprotein in the clearance of the Alzheimer’s disease Aβ1-42 peptides. Cell Stress & Chaperones. 2015; 20: 421–430.
[126]
Hartz AMS, Zhong Y, Shen AN, Abner EL, Bauer B. Preventing P-gp Ubiquitination Lowers Aβ Brain Levels in an Alzheimer’s Disease Mouse Model. Frontiers in Aging Neuroscience. 2018; 10: 186.
[127]
Chiu C, Miller MC, Monahan R, Osgood DP, Stopa EG, Silverberg GD. P-glycoprotein expression and amyloid accumulation in human aging and Alzheimer’s disease: preliminary observations. Neurobiology of Aging. 2015; 36: 2475–2482.
[128]
Wang W, Bodles-Brakhop AM, Barger SW. A Role for P-Glycoprotein in Clearance of Alzheimer Amyloid β -Peptide from the Brain. Current Alzheimer Research. 2016; 13: 615–620.
[129]
Feng M, Zhou Q, Xie H, Liu C, Zheng M, Zhang S, et al. Role of CD36 in central nervous system diseases. Neural Regeneration Research. 2024; 19: 512–518.
[130]
Silverstein RL, Febbraio M. CD36, a scavenger receptor involved in immunity, metabolism, angiogenesis, and behavior. Science Signaling. 2009; 2: re3.
[131]
Febbraio M, Hajjar DP, Silverstein RL. CD36: a class B scavenger receptor involved in angiogenesis, atherosclerosis, inflammation, and lipid metabolism. The Journal of Clinical Investigation. 2001; 108: 785–791.
[132]
Li J, Li M, Ge Y, Chen J, Ma J, Wang C, et al. β-amyloid protein induces mitophagy-dependent ferroptosis through the CD36/PINK/PARKIN pathway leading to blood-brain barrier destruction in Alzheimer’s disease. Cell & Bioscience. 2022; 12: 69.
[133]
Park L, Wang G, Zhou P, Zhou J, Pitstick R, Previti ML, et al. Scavenger receptor CD36 is essential for the cerebrovascular oxidative stress and neurovascular dysfunction induced by amyloid-beta. Proceedings of the National Academy of Sciences of the United States of America. 2011; 108: 5063–5068.
[134]
Miao L, St Clair DK. Regulation of superoxide dismutase genes: implications in disease. Free Radical Biology & Medicine. 2009; 47: 344–356.
[135]
Kuroda J, Ago T, Nishimura A, Nakamura K, Matsuo R, Wakisaka Y, et al. Nox4 is a major source of superoxide production in human brain pericytes. Journal of Vascular Research. 2014; 51: 429–438.
[136]
Špiranec K, Chen W, Werner F, Nikolaev VO, Naruke T, Koch F, et al. Endothelial C-Type Natriuretic Peptide Acts on Pericytes to Regulate Microcirculatory Flow and Blood Pressure. Circulation. 2018; 138: 494–508.
[137]
Schultz N, Nielsen HM, Minthon L, Wennström M. Involvement of matrix metalloproteinase-9 in amyloid-β 1-42-induced shedding of the pericyte proteoglycan NG2. Journal of Neuropathology and Experimental Neurology. 2014; 73: 684–692.
[138]
Arihiro S, Ohtani H, Hiwatashi N, Torii A, Sorsa T, Nagura H. Vascular smooth muscle cells and pericytes express MMP-1, MMP-9, TIMP-1 and type I procollagen in inflammatory bowel disease. Histopathology. 2001; 39: 50–59.
[139]
Stomrud E, Björkqvist M, Janciauskiene S, Minthon L, Hansson O. Alterations of matrix metalloproteinases in the healthy elderly with increased risk of prodromal Alzheimer’s disease. Alzheimer’s Research & Therapy. 2010; 2: 20.
[140]
Qu L, Ji L, Wang C, Luo H, Li S, Peng W, et al. Synthesis and evaluation of multi-target-directed ligands with BACE-1 inhibitory and Nrf2 agonist activities as potential agents against Alzheimer’s disease. European Journal of Medicinal Chemistry. 2021; 219: 113441.
[141]
Moussa C, Hebron M, Huang X, Ahn J, Rissman RA, Aisen PS, et al. Resveratrol regulates neuro-inflammation and induces adaptive immunity in Alzheimer’s disease. Journal of Neuroinflammation. 2017; 14: 1.
[142]
Kimura T, Hashimura T, Miyakawa T. Observations of microvessels in the brain with Alzheimer’s disease by the scanning electron microscopy. The Japanese Journal of Psychiatry and Neurology. 1991; 45: 671–676.
[143]
Hibbs E, Love S, Miners JS. Pericyte Contractile Responses to Endothelin-1 and Aβ Peptides: Assessment by Electrical Impedance Assay. Frontiers in Cellular Neuroscience. 2021; 15: 723953.
[144]
Briyal S, Ranjan AK, Gulati A. Oxidative stress: A target to treat Alzheimer’s disease and stroke. Neurochemistry International. 2023; 165: 105509.
[145]
Lourenco MV, Frozza RL, de Freitas GB, Zhang H, Kincheski GC, Ribeiro FC, et al. Exercise-linked FNDC5/irisin rescues synaptic plasticity and memory defects in Alzheimer’s models. Nature Medicine. 2019; 25: 165–175.
[146]
Fukushi JI, Makagiansar IT, Stallcup WB. NG2 proteoglycan promotes endothelial cell motility and angiogenesis via engagement of galectin-3 and alpha3beta1 integrin. Molecular Biology of the Cell. 2004; 15: 3580–3590.
[147]
Nielsen HM, Ek D, Avdic U, Orbjörn C, Hansson O, Netherlands Brain Bank, et al. NG2 cells, a new trail for Alzheimer’s disease mechanisms? Acta Neuropathologica Communications. 2013; 1: 7.
[148]
Van den Steen PE, Dubois B, Nelissen I, Rudd PM, Dwek RA, Opdenakker G. Biochemistry and molecular biology of gelatinase B or matrix metalloproteinase-9 (MMP-9). Critical Reviews in Biochemistry and Molecular Biology. 2002; 37: 375–536.
[149]
Anwar MM, Özkan E, Gürsoy-Özdemir Y. The role of extracellular matrix alterations in mediating astrocyte damage and pericyte dysfunction in Alzheimer’s disease: A comprehensive review. The European Journal of Neuroscience. 2022; 56: 5453–5475.
[150]
Maia LF, Mackenzie IRA, Feldman HH. Clinical phenotypes of Cerebral Amyloid Angiopathy. Journal of the Neurological Sciences. 2007; 257: 23–30.
[151]
Rabin JS, Nichols E, La Joie R, Casaletto KB, Palta P, Dams-O’Connor K, et al. Cerebral amyloid angiopathy interacts with neuritic amyloid plaques to promote tau and cognitive decline. Brain: a Journal of Neurology. 2022; 145: 2823–2833.
[152]
Verbeek MM, Van Nostrand WE, Otte-Höller I, Wesseling P, De Waal RM. Amyloid-beta-induced degeneration of human brain pericytes is dependent on the apolipoprotein E genotype. Annals of the New York Academy of Sciences. 2000; 903: 187–199.
[153]
Li Y, Xiao D, Wang X. The emerging roles of ferroptosis in cells of the central nervous system. Frontiers in Neuroscience. 2022; 16: 1032140.
[154]
Hallmann R, Horn N, Selg M, Wendler O, Pausch F, Sorokin LM. Expression and function of laminins in the embryonic and mature vasculature. Physiological Reviews. 2005; 85: 979–1000.
[155]
Parodi-Rullán R, Ghiso J, Cabrera E, Rostagno A, Fossati S. Alzheimer’s amyloid β heterogeneous species differentially affect brain endothelial cell viability, blood-brain barrier integrity, and angiogenesis. Aging Cell. 2020; 19: e13258.
[156]
Iadecola C, Zhang F, Niwa K, Eckman C, Turner SK, Fischer E, et al. SOD1 rescues cerebral endothelial dysfunction in mice overexpressing amyloid precursor protein. Nature Neuroscience. 1999; 2: 157–161.
[157]
Zhu L, Lu F, Zhang X, Liu S, Mu P. SIRT1 Is Involved in the Neuroprotection of Pterostilbene Against Amyloid β 25-35-Induced Cognitive Deficits in Mice. Frontiers in Pharmacology. 2022; 13: 877098.
[158]
Palmer J, Love S. Endothelin receptor antagonists: potential in Alzheimer’s disease. Pharmacological Research. 2011; 63: 525–531.
[159]
Elesber AA, Bonetti PO, Woodrum JE, Zhu XY, Lerman LO, Younkin SG, et al. Bosentan preserves endothelial function in mice overexpressing APP. Neurobiology of Aging. 2006; 27: 446–450.
[160]
Dumont AS, Dumont RJ, McNeill JH, Kassell NF, Sutherland GR, Verma S. Chronic endothelin antagonism restores cerebrovascular function in diabetes. Neurosurgery. 2003; 52: 653–660; discussion 659–660.
[161]
Rodriguez-Porcel M, Lerman LO, Holmes DR, Jr, Richardson D, Napoli C, Lerman A. Chronic antioxidant supplementation attenuates nuclear factor-kappa B activation and preserves endothelial function in hypercholesterolemic pigs. Cardiovascular Research. 2002; 53: 1010–1018.
[162]
Ruopp NF, Cockrill BA. Diagnosis and Treatment of Pulmonary Arterial Hypertension: A Review. JAMA. 2022; 327: 1379–1391.
[163]
Yan SS, Chen D, Yan S, Guo L, Du H, Chen JX. RAGE is a key cellular target for Abeta-induced perturbation in Alzheimer’s disease. Frontiers in Bioscience (Scholar Edition). 2012; 4: 240–250.
[164]
Lee YS, Kim H, Kim YH, Roh EJ, Han H, Shin KJ. Synthesis and structure-activity relationships of tri-substituted thiazoles as RAGE antagonists for the treatment of Alzheimer’s disease. Bioorganic & Medicinal Chemistry Letters. 2012; 22: 7555–7561.
[165]
Galasko D, Bell J, Mancuso JY, Kupiec JW, Sabbagh MN, van Dyck C, et al. Clinical trial of an inhibitor of RAGE-Aβ interactions in Alzheimer disease. Neurology. 2014; 82: 1536–1542.
[166]
Sabbagh MN, Agro A, Bell J, Aisen PS, Schweizer E, Galasko D. PF-04494700, an oral inhibitor of receptor for advanced glycation end products (RAGE), in Alzheimer disease. Alzheimer Disease and Associated Disorders. 2011; 25: 206–212.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top