v
Search
Advanced Search

Publications > Journals > Exploratory Research and Hypothesis in Medicine > Article Full Text

  • OPEN ACCESS

Pericytes and the Neurovascular Unit: The Critical Nexus of Alzheimer Disease Pathogenesis?

  • Steven P. Cercy* 
 Author information
Exploratory Research and Hypothesis in Medicine   2021;6(3):125-134

doi: 10.14218/ERHM.2020.00062

Abstract

Alzheimer disease (AD) has been viewed as the quintessential neurodegenerative disorder, and has defied decades of extensive research to find safe and effective disease-modifying treatment approaches. However, over the last 15–20 years, a new focus has developed on the role of vascular dysfunction in AD. Key to this approach is the consideration of the non-neuronal cells and other structural elements comprising the neurovascular unit (NVU), in particular pericytes. This review will examine the role of pericytes and the NVU in AD pathogenesis and the manner in which they interact with traditional factors, such as neuroinflammation, amyloid-beta, and apolipoprotein E. Based on the emerging evidence of the unique properties of pericytes, these “forgotten cells” might represent a crucial nexus for solving the mysteries of AD.

Keywords

Pericyte, Neurovascular unit, Blood-brain barrier, Alzheimer disease, Neurodegenerative disorders, Amyloid-beta, Apolipoprotein E

Introduction

Alzheimer disease (AD) is the pre-eminent enigma in clinical neuroscience. The disorder was first described in 1907 by the German psychiatrist and neuropathologist Alois Alzheimer.1,2 To date, AD has resisted sustained efforts to develop effective disease-modifying therapies. From the first-generation cholinesterase inhibitor, tacrine,3 to second-generation agents, including donepezil4 and memantine,5 to the development of monoclonal antibodies, such as solanezumab6 and aducanumab,7 investigators have failed to achieve satisfactory clinical endpoints. Some therapies have proved to be dangerous and serious adverse events have been observed, such as hepatotoxicity,8 meningoencephalitis,9 and cerebral edema.10

From its initial description, AD has been conceptualized as representing the quintessential, pure neurodegenerative disorder. However, this perspective of AD belies its complexities and represents one of the false dichotomies that remain in the neurosciences. The most notable of these might be the outdated (yet still widely taught in medical schools and residency programs) distinction between “organic” versus “functional” disorders in neurology and psychiatry. As neuroscience researchers have expanded the knowledge base, the array of disordered functions at the molecular, cellular, behavioral, and cognitive levels has been revealed.

The previous view of AD pits it against vascular dementia (VD). Diagnostic classification systems of dementia syndromes have traditionally assumed that a clear and reliable distinction exists between AD and VD. In recognition of its own complexities, the conceptualization of VD has undergone revision. The original terminology—multi-infarct dementia11—was changed to its current and more inclusive nosology, which reflects the fact that small-vessel disease contributes more frequently to cognitive decline than the accrual of large-vessel infarctions.12 Throughout these changes in thinking and terminology, VD has continued to be viewed as the prime example of dementia caused by vascular pathology.

Accumulating evidence has compelled researchers and clinicians to reconcile these findings and develop more refined and comprehensive hypotheses. It is now understood that the pathophysiology of AD is not limited to the classical view of neuronal degeneration. It also involves the full complement of glia, and cells that comprise the cerebral vasculature and other structural components, which are collectively referred to as the neurovascular unit (NVU). This review presents the evolution of knowledge on AD pathogenesis, beginning with the initial identification of the principal risk factor for sporadic AD, the discovery of a range of autosomal dominant mutations that cause familial AD, and the insights obtained from those rare cases. Then, it will discuss the increased prominence of the role of vascular mediators for AD and consider the specific processes through which the new data on the blood-brain barrier (BBB), pericytes, and other components of the NVU could provide a basis to integrate previously disparate factors into a comprehensive modern viewpoint.

The classical view of Alzheimer disease pathophysiology: background and context

Factors that were initially identified as principal contributors to AD pathophysiology were amyloid-beta (Aβ) protein, tau protein, and apolipoprotein E (APOE). Each will be considered individually. APOE was the first established risk-mediating variable associated with sporadic AD.13 In particular, the ε4 allele of APOE (APOE4) confers an elevated risk of sporadic AD. APOE4 operates dose-dependently. Heterozygosity for ε4 results in a 2–5 fold increased risk for the development of AD; ε4 homozygosity increases AD risk by approximately 5–10 fold.13–15 In contrast, the APOE ε2 allele is protective, whereas the ε3 allele is neutral for AD risk.

Despite the evidence that APOE confers substantial risk of AD, questions remained regarding the nature of its contribution to AD pathogenesis until recently. Ultimately, investigators determined that APOE interacts with Aβ by functioning as a chaperone or catalytic protein. APOE2 inhibits the polymerization of Aβ monomers to form toxic oligomeric species but APOE4 promotes β polymerization,16,17 findings that are consistent with epidemiological studies of AD risk. In addition, APOE is an important component that regulates the clearance of Aβ from the extracellular space. The APOE4 variant is the least efficient at clearing Aβ.18

Around the time that the importance of APOE was recognized, advances in genetic sequencing analyses allowed for the rapid identification and characterization of mutations associated with clinical presentations of early-onset familial AD (EOFAD). The first was identified in 1991, affecting amyloid precursor protein (APP), and accounts for approximately 15%–20% of all EOFAD cases.19 The gene for APP resides on chromosome 21. Overexpression of APP explains the high incidence of AD among individuals with trisomy 21Down syndrome—due to the extra chromosome.20 Symptom onset typically occurs in the late fourth to fifth decade of life among people affected by Down syndrome, which reflects the dose-dependent nature of APP translation.

APP is a single-pass transmembrane protein that is involved in synapse production and maintenance and cellular signaling.21 APP undergoes sequential cleavage by the gamma (γ)-secretase complex, which yields a specific set of polypeptide products.22 The most important is Aβ, one of the principal markers of AD pathology. When APP undergoes γ-secretase processing, Aβ monomers are released primarily into the extracellular space. Aggregation of Aβ begins when amyloidogenic monomers attract and attach to additional colocated Aβ monomers to form oligomers. Aβ oligomers appear to be the most toxic form of Aβ, particularly the production of 40- and 42-residue isoforms via the amyloidogenic pathway. In addition to their propensity to aggregate, Aβ oligomers exert direct neurotoxic effects.23

Accumulation of oligomers leads progressively to the production of insoluble Aβ fibrils, which self-propagate to form plaques24 that are highly resistant to proteolysis and clearance. Regions of Aβ seeding then continue to propagate along interconnected neuroanatomic pathways,25 in a manner similar to the process that occurs in prion diseases.26 Aβ fibrils and mature plaques induce an inflammatory response that is mediated by astrocytes and activated microglia, which generate reactive oxygen species (ROS) and leads to oxidative stress and apoptosis.27 In addition to increased production, dysfunctional clearance of Aβ has been implicated in AD pathophysiology.28 Aβ clearance occurs via several mechanisms: glial endocytosis, proteolytic enzymatic degradation, transport across the BBB mediated by low-density lipoprotein family receptors, activation of the complement arm of the immune response, and passive drainage through interstitial perivascular spaces and specialized lymphatic vessels, referred to as the glymphatic system.29,30 Events subsequent to Aβ deposition and aggregation—neuroinflammation, generation of ROS, excitotoxicity, tau hyperphosphorylation, microtubule disruption, and ultimately neuronal apoptosis—are referred to as the amyloid cascade hypothesis (ACH).

The ACH initially came under criticism when studies showed that the correlation between cerebral Aβ burden and cognitive function was weak31 and when evidence of neuronal injury was observed to precede Aβ deposition, such as in a murine model that over-expresses Aβ.32 These findings raised doubts that the ACH was sufficient to account for AD pathophysiology. Accordingly, increased attention turned toward the other major marker of neuropathology in AD, tau protein. Microtubules are composed principally of the protein tubulin; together with tau, these proteins form a critical structural component of neurons. Tau is a soluble phosphoprotein that acts to stabilize microtubules,33 and performs several other important functions which include providing intracellular axonal transport, regulating synaptic plasticity, and supporting the structural integrity of intraneuronal signaling pathways.34

Under pathological conditions, tau becomes hyperphosphorylated, which decreases the affinity of tau toward tubulin and leads to the dissociation of tau from microtubules, thereby resulting in their destabilization and the formation of insoluble tau aggregates. These events contribute to structural degradation and lead to neuronal death.35 The characteristic pathological marker of tau dysfunction is the flame-shaped neurofibrillary tangle that, when co-located with aggregations of Aβ, are referred to as neuritic plaques. In contrast to the weak relationship between Aβ levels and cognitive impairment, tau density is strongly correlated with dementia staging.36

Subsequent research revealed that, although tau protein is an important mediator of AD pathogenesis, Aβ was a necessary condition for the development of the cognitive syndrome of AD.37 For example, when Aβ was absent, there was no association between tau binding and hippocampal volume. In contrast, in the presence of Aβ, tau binding was greater and was associated with lower hippocampal volume.38 Apart from modifications to the ACH, investigators believed that other more fundamental factors contributing to AD pathogenesis were likely to play an important role.

Relationships between Alzheimer disease and vascular risk

The early conceptualization of AD focused heavily on the neurodegenerative aspects of its pathophysiology. In addition, cerebrovascular disease (CVD) and related dementia syndromes, as exemplified by VD, were viewed as occupying two poles of a single spectrum, with neurodegeneration at one end and vascular pathology at the other. However, overlaps between AD and CVD have been uncovered; among the earliest research to demonstrate an overlap was the Rotterdam study.39 Of note, Alois Alzheimer found1 that neurofibrillary tangles co-existed with cerebral microvascular arteriosclerotic disease,40 the first clue into the role of vascular pathology in neurodegeneration.

Several factors identified as promoting risk for CVD have been shown to confer risk for AD. These include hypertension, hyperlipidemia, type 2 diabetes, and cigarette smoking.41–45 These shared risk variables raised new questions about the processes that underlie the generation of AD pathology. This view coincided with an increased awareness of the relevance of changes that involve the cerebral microvasculature in AD,42,46–47 and suggested that rather than existing as discrete entities, neurodegeneration and microvascular disease constituted two ends of a common spectrum of pathology.48 Therefore, research into factors that mediate vascular damage in AD began.

The most direct relationship between AD-type pathology and CVD manifests as an intramural deposition of Aβ within cerebral arteries, arterioles, and capillaries, as well as meninges, a condition that is called cerebral amyloid angiopathy (CAA).49 The functional effect of Aβ infiltration is a weakening of vessel walls, which makes them susceptible to leakage, results in microhemorrhage,50 and less frequently, rupture causing large-vessel hemorrhage. Accordingly, CAA is a major risk factor for stroke.51 Although CAA and AD have overlapping features, they are considered distinct diagnostic entities. As with AD, most CAA cases are sporadic. Increased production and diminished clearance of Aβ have been implicated as underlying mechanisms of sporadic CAA.29 CAA and AD share APOE4 as the primary risk factor for sporadic disease.

However, there are important distinctions between these diseases.52 Aβ40 is the predominant species deposited in CAA as opposed to Aβ42, which dominates in AD. In addition, APOE2 is protective in AD, but it is associated with an enhanced risk of blood vessel breakdown in CAA. Familial, or hereditary CAA (HCAA) is caused by a variety of rare autosomal dominant mutations. Six HCAA variants have been identified to date, and only the Dutch-type is caused by a mutation to APP on chromosome 21, as in familial AD. All other HCAA variants consist of mutations that code for amyloid proteins other than Aβ.

The BBB and neuroinflammation

The BBB is composed of specialized endothelial cells that form the walls of all cerebral vessels. These cells are interconnected by tight junctions that strictly limit permeability bidirectionally and serve to compartmentalize the parenchyma from the blood. The BBB is a component of a larger, integrated set of elements that comprise the NVU, which will be discussed in the following sections.

Dogma once held that the brain was immune-privileged; that is, it was assumed that innate and adaptive immune system components were sequestered from the brain by the BBB. However, the BBB is neither impenetrable nor impervious as previously understood. B lymphocytes migrate from the periphery across the BBB,53 where they become activated and perform immune regulating functions within the central nervous system (CNS).54 These activated microglia secrete interleukin-6 (IL-6) within the CNS compartment.55 In addition to local cytokine production, circulating IL-1α and tumor necrosis factor-alpha (TNF-α)46,56 enter the CNS via active transport.

Complex links between neuroinflammation and BBB function have been discovered. Endothelial cells secrete pro-inflammatory cytokines. When exposed to Aβ40 species in vitro, cultured human brain endothelial cells respond by up-regulating gene expression for inflammatory cytokines IL-1β and IL-6.57 Endothelial cells are primary regulators of Aβ influx into the brain58 via receptors for advanced glycation end products (RAGE),59 a process that contributes to the propagation of the inflammatory response. Pro-inflammatory cytokines IL-2 and IL-6,60,61 regulate BBB permeability. Evidence indicates that age-related BBB disruption is more pronounced among individuals with cognitive dysfunction.62 Similarly, levels of IL-1β, IL-6, and TNF-α in endothelial cells are higher in AD patients than in cognitively intact individuals.63

Pericytes and the neurovascular unit: overview

The NVU consists of three major components: neurons, glia, and vascular cells. The vascular cells and their interactions with the other components have been the target of recent research. In addition to the endothelial cells, the basal lamina (also referred to as the basement membrane), microvascular smooth muscle cells, and pericytes comprise the vascular component of the NVU. The endothelium forms the first structural layer of the NVU and is in direct contact with plasma and other blood components. The second cellular layer of the NVU consists of the end feet of astrocytes, the basal membrane, which is an extension of astrocyte end feet, and the pericytes. Perictyes envelop cerebral capillaries, and precapillary arterioles and venules, and make anatomical contact with endothelial cells (Fig. 1).64 A host of disturbances that involve pericytes have been shown to occur during physiological aging. Disturbances of pericyte function might represent a unifying feature of disparate lines of evidence that accounts for age-related neuronal loss, neurodegeneration, and the pathophysiology of AD (Fig. 2).

Schematic representation of the neurovascular unit.
Fig. 1  Schematic representation of the neurovascular unit.64

Penetrating arteriole branches into arterioles and capillaries, which then drain via venules into veins, returning to subarachnoid space. Schematic cross sections of arterial, capillary, and venous levels are shown, each with its vessel-associated cell types. The box depicts how pericytes establish direct connections with endothelial cells through peg-socket contacts. AEF, astrocyte endfoot; BM, basement membrane; EC, endothelial cell; PC, pericyte; PVF, perivascular fibroblast; SMC, smooth muscle cell. Source: Lendahl et al. 2019, Figure © EMBO. Reproduced under the terms of the Creative Commons Attribution 4.0 License which permits use, distribution, and reproduction in any medium, provided the original work is properly cited.

Idealized longitudinal cross section of the neurovascular unit summarizing molecular mechanisms of pericyte regulation and their relationships with AD pathophysiology.
Fig. 2  Idealized longitudinal cross section of the neurovascular unit summarizing molecular mechanisms of pericyte regulation and their relationships with AD pathophysiology.

Microglia release ROS and inflammatory factors in the presence of Aβ aggregates. Accumulating extracellular Aβ is transported into endothelial cells and perictyes via RAGE, triggering transcription factors, such as NF-κB and MMP-9 to produce IL-x and TNF-α, setting up a feed-forward loop of accelerating Aβ production and neuroinflammation. APOE4 secreted by astrocytes is taken up by perictyes via LRP-1 and further promotes release of inflammatory factors, but APOE2 and APOE3 inhibit that pathway. Aβ induces a significant increase in NOX4 that inhibits pericyte proliferation and downstream angiogenesis. Aβ interferes directly with tight junction proteins, increasing BBB permeability. ET-1 secreted by endothelial cells binds to ETA receptors on perictyes, causing them to contract, leading to capillary constriction. Hyperglycemic states inhibit the interaction between PDGF secreted by endothelial cells and its receptor PGDFR-β found on perictyes, contributing to pericyte apoptosis, endothelial proliferation, capillary remodeling and BBB breakdown. Aβ, amyloid beta; AEF, astrocyte endfoot; APOEx, apolipoprotein Ex; BBB, blood-brain barrier; EC, endothelial cell; ET-1, endothelin-1; ETA, ET-1 type A receptor; HPN, hippocampal pyramidal neuron; IL-x, interleukin-x, LPR-1, low-density lipoprotein receptor-related protein-1; MG, microglial cell; MMP-9, matrix metalloproteinase-9 complex; NF-κB, nuclear-factor kappa B; NOX4, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4; PC, pericyte; PDGF, platelet-derived growth factor; PDGFR-β, platelet-derived growth factor receptor-beta; RAGE, receptor for advanced glycation end products; ROS, reactive oxygen species; TJ, tight junction; TNF-α, tumor necrosis factor-alpha; VEGF, vascular endothelial growth factor. Figure © Steven P. Cercy.

Pericytes and aging

One consequence of physiological aging is the increased production and diminished scavenging of various ROS, in particular, the free radical superoxide anion (O2). Increased ROS levels affect many organ systems, including the cerebral microvasculature. The accumulation of ROS species gradually leads to mitochondrial dysfunction, DNA damage, and apoptosis. In addition, O2 breaks down nitric oxide (NO), which is a regulator of vascular tone in its role as a vasodilator. Aging is associated with reduced bioavailability of NO in multiple organ systems.65 Perictytes are among the cells that are subject to the cumulative deleterious effects of age-associated ROS overexpression.

Aging pericytes develop certain deviations of their ultrastructural elements; these include intracellular inclusions, pinocytotic vesicles, enlarged lipid granules, and mitochondrial abnormalities, all of which suggest cellular dysfunction, degeneration, or both.66 Alterations of desmin protein filaments in pericytes have been observed,67 which suggests a disturbance of their cellular structure. In human elders, pericytes become depopulated68 and show a significant reduction in their area of capillary coverage.69

The precise roles that pericytes play under optimal physiological conditions and during normal aging have not yet been fully elucidated. Research over the past 10–15 years, however, has revealed several key functions that these cells perform during the development and maintenance of cerebral microcirculation. Pericytes control microvascular blood flow directly via contraction, which causes capillary constriction70 and, under pathological conditions, ischemia which might lead to localized hypoxemia.71 They play an important role in angiogenesis72; associated properties include migration and variability in phenotype, alignment, and endothelial cell contact. Pericytes guide and determine the direction and branching of newly formed blood vessels,73 prevent vessel regression,74 and promote endothelial cell survival.75 Pericytes in the hypothalamus serve an important role in the regulation of glucose levels through insulin signaling. In a murine model, perictyes increased insulin sensitivity in hypothalamic neurons in a dose-dependent fashion; neither astrocytes nor vascular smooth muscle cells contributed to that process.76 This finding suggests that hypothalamic pericyte loss might be implicated specifically in the dysregulation of insulin sensitivity, which is a fundamental aspect of diabetes.

The molecular mechanisms that drive the formation and maintenance of the cerebral microvasculature are centered on intracellular signaling. Platelet-derived growth factor (PDGF) is secreted by endothelial cells and binds to platelet-derived growth factor receptor-beta (PDGFR-β), which is expressed by pericytes. This ligand-receptor complex activates signal transduction pathways that regulate migration and proliferation of pericytes toward endothelial cells that compose the vascular wall.77 Experimental models of diabetic retinopathy that used pericyte-deficient mice revealed that chronic hyperglycemia resulted in diminished PDGFR-β signaling, which leads to pericyte apoptosis.78 Pericyte loss induces endothelial cell proliferation with increased numbers of abnormal acellular capillaries,74,79 rather than endothelial apoptosis.80 Downstream events that are related to pericyte changes include significant capillary remodeling, which is characterized by vessel dilation and tortuosity, as well as basal lamina hypertrophy.67,81

Subsequent studies revealed the particular importance of pericytes in microvascular regulation; they are the only cells of the NVU that express PDGFR-β to enable a response to PDGF.80 Moreover, soluble PDGFR-β has been identified as a specific marker of pericyte injury.82 In addition to PDGF, vascular endothelial growth factor is secreted by pericytes under hypoxic conditions, which stimulates proliferation and migration of additional pericytes.83 Nuclear factor-kappa B (NF-κB), an important transcription factor that mediates the inflammatory response, is activated in a subpopulation of pericytes in response to exercise, which promotes angiogenesis.84

In concert with their angiogenic properties, pericytes are crucial to maintain the integrity of the BBB. Vascular remodeling that occurs with pericyte apoptosis contributes to capillary destabilization,67 which causes the breakdown of the BBB. Pericytes regulate the formation of endothelial tight junctions that modulate vascular permeability. Pericyte structural deterioration and apoptosis via any mechanism results in reduced contact with and coverage of endothelial cells67,85 and the breakdown of the BBB, consequently permitting the influx and accumulation of serum proteins including the key coagulation macromolecules thrombin, fibrinogen, and fibrin—which thrombin cleaves from fibrinogen—as well as plasmin and hemoglobin.80

Elevated thrombin in AD86 contributes to neuronal loss, vascular injury, and cognitive impairment.87 Fibrinogen exacerbates the neurotoxic effects of Aβ.88 Moreover, fibrinogen leakage from injured cerebral microvessels activates resident microglia bearing CD-11b receptors, which then prune neuronal dendritic spines and whole dendrites.89 Protein influx from all sources contributes to cerebral edema that might cause further capillary compression.90 The NF-κB signaling pathway is a fundamental link between thrombosis and inflammation.91 Free iron from degraded hemoglobin generates ROS.92 All of these aberrant processes contribute to secondary neuronal degeneration.90,93

Pericytes and AD pathophysiology

Based on the foregoing research findings, the issues to be determined are to what extent pericyte dysfunction contributes to or interacts with classical AD pathophysiological processes, and by what mechanisms those interactions occur. Pericyte degeneration begins early in AD, particularly in hippocampal neurons, which are among the first cells involved in AD pathology. Reductions in pericyte coverage correlate inversely with evidence of BBB permeability,62 as measured by hippocampal levels of plasma proteins, including immunoglobulin G and fibrin.94 Elevated soluble PDGFR-β in cerebrospinal fluid is an early biomarker of cognitive dysfunction, which is independent of Aβ and tau levels.82,95 This shows that pericyte-specific dysfunction, which is characterized by vascular regression and disrupted vascular permeability, is associated with key attributes of AD pathology.93

Pericytes, APOE and Aβ

As APOE and Aβ functions have been further elucidated, their relationships with pericytes and other elements of the NVU have come into focus. APOE2 and APOE3 are secreted by astrocytes and taken up by pericytes via low-density lipoprotein receptor-related protein-1 (LRP-1), where they inhibit a key pro-inflammatory pathway, the cyclophilin A-nuclear factor B-matrix metalloproteinase 9 complex. However, APOE4 promotes that pathway,66,96 which directly increases pericyte injury97 and impairs the formation of basement membranes.98 Thus, APOE4 accelerates pericyte loss relative to carriers of APOE2 and APOE3 alleles.

It is well known that in AD, cerebral capillary constriction is provoked by Aβ via the production of ROS.99 However, the mechanisms of this and locus of action remained unclear. Aβ enhances the activity of nicotinamide adenine dinucleotide phosphate oxidases (NOXs) to form reactive superoxides. NOX4 is found in pericytes and endothelial cells. In a transgenic mouse model, Aβ increased NOX4 levels sevenfold, and the blockade of NOX4 prevented capillary constriction following the application of Aβ. In contrast, NOX2, which is found in macrophages, increased by only twofold in the presence of Aβ, and blocking NOX2 produced a substantially attenuated effect on capillary relaxation. Similar findings were elicited using human brain slices treated using comparable methods.89

RAGE, which is expressed by neurons, glia, vascular smooth muscle cells, endothelial cells, and pericytes, has a crucial role in the influx of peripheral Aβ into the parenchyma. RAGEs bind to Aβ and a broad range of compounds that are referred to as advanced glycation end products (AGEs), also known as glycotoxins. These compounds are produced as a normal consequence of lipid and protein metabolism and are present in foods that are cooked at high temperatures. During physiological aging, AGEs accumulate progressively within all cells. However, in AD and diabetes, this normal process is accelerated. AGEs are present within Aβ plaques and neurofibrillary tangles of patients with AD. Accumulated AGEs contribute to the induction of oxidative stress by glial cells.100 Increases in RAGE protein and RAGE-expressing microglia occur in AD, and correlate with disease severity.101

Pericytes are crucial for regulating Aβ trafficking between the BBB and parenchyma via the LRP-1 and RAGE pathways described previously. They assist with Aβ clearance by phagocytosis and promote Aβ efflux from parenchyma via the LRP-1 pathway.102 Despite this role, pericytes remain susceptible to Aβ toxicity.103 In AD, the accumulation of Aβ within pericytes leads to dysmorphic remodeling104 and apoptosis. In a transgenic model of pericyte-deficient viable mice overexpressing APP, degeneration and loss of pericytes resulted in elevated levels of Aβ40 and Aβ42 in the brain, as well as increased levels of tau not typically observed in this transgenic model. Reinforcement of this destructive cycle occurs when reduced cerebral blood flow leads to further synthesis and accelerating burden of Aβ, worsening pericyte apoptosis, progressive microvascular injury, and cognitive impairment.102

Endothelin-1 (ET-1), which is a powerful vasoconstrictor that is secreted by endothelial cells, also appears to play an important role in Aβ-mediated vascular insult. Levels of ET-1 are elevated in individuals with AD, and are up-regulated by Aβ.105 Subsequent research revealed that Aβ oligomers induced the release of ROS in pericytes, which enhances transcription and release of ET-1,106 which then binds ET-1 type A (ETA) receptors. It is this ligand-receptor complex that causes pericytes to contract, which leads to capillary constriction, and results in hypoxic ischemia, local hypoglycemia, and neuronal loss in affected microregions. Constriction of capillaries worsens with increasing Aβ load.107 Furthermore, BBB disruption is directly related to the influx of circulating Aβ into brain parenchyma.94

The intramural deposition of Aβ within the walls of the cerebral vasculature that occurs in CAA is an important yet overlooked component of AD pathology.108 Cultured human brain pericytes exposed in vitro to the Dutch-type mutation that affects the Aβ40 species is the predominant species that induces the formation of Aβ fibrils at the cell surface, which leads to pericyte degeneration. Insulin appears to inhibit Aβ fibril formation in a dose-dependent manner, further suggesting that insulin might be involved in the regulation of Aβ fibrillization and might mitigate Aβ-induced pericyte loss in AD.109 In humans with CAA, dystrophic neurites are found within the perivascular spaces of capillaries in occipital association cortex, more so than in primary occipital cortex. In addition, the density of dystrophic neurites correlates with Aβ levels.108 Relationships among insulin regulation, Aβ fibril formation, and the deposition of Aβ in intramural and perivascular spaces have implications for diabetes and the risk it poses for the development of sporadic AD.

Aβ interferes directly with the integrity of endothelial tight junctions, which are regulated by pericytes and leads to an inappropriate increase in BBB permeability.60 In addition, Aβ stabilizes fibrin clots;110 increased fibrin deposition has been observed in vessels that are affected by CAA.74 Of note, age-dependent vascular damage in pericyte-deficient mice precedes neuroinflammation, neurodegeneration, and consequent impairments in learning and memory.80 This is consistent with cerebral hypometabolism outweighing the degree of volume loss among humans who are presymptomatic EOFAD mutation carriers.111

Pericytes and tau

In addition to the deleterious influence of Aβ on the BBB that was identified recently, tau plays a significant but less understood role in the regulation of BBB integrity. In a transgenic mouse model, age-related increases in tau were associated with increased BBB permeability against erythrocytes, peripheral T lymphocytes, and immunoglobulin G. This effect was first observed in the hippocampus,112 where the earliest tau aggregation occurs in AD. The migration of cells and molecules of the immune system, which is facilitated by a leaky BBB, helps to drive neuroinflammation.113 However, detectable neuroinflammation and neurodegeneration do not occur until well after the initiation of BBB deterioration.112 One mechanism through which tau might influence BBB permeability occurs through its interactions with tight junctions and adherens junctions between endothelial cells and their actin cytoskeleton proteins. Disrupting this association appears to result in tau-induced neurotoxicity.114 Pericyte deficiency in a transgenic model of AD in mice led to increased phosphorylation of tau in the hippocampus and cortex.102 Collectively, these processes could potentially cause a synergistic effect between pericyte loss, tau generation, Aβ accumulation, and neurodegeneration.

Future directions

Based on recent and emerging evidence, pericytes present a novel opportunity to shift the focus of disease-modifying therapies away from failed efforts to break the link between amyloid deposition and cognitive decline. AD pathology appears to develop 15–20 years prior to symptom onset. One likely reason for the failure of available therapies could be that the development of pathology in AD has advanced too far to be amenable to treatment once individuals become symptomatic. Thus, the main thrust of AD research over the last decade has been to identify early markers of AD well in advance of symptom onset.115

One potential approach entails a renewed focus on neuroinflammation. However, rather than using traditional NSAIDs, inflammation that is mediated by IL-1β, IL-6, and other pro-inflammatory cytokines could be specifically targeted in an attempt to block Aβ-mediated generation of ROS in pericytes.116 The Aβ-RAGE-cyclophilin A-nuclear factor B-matrix metalloproteinase 9 cascade, which is an important mechanism of AD pathogenesis via BBB disruption might be a promising potential target for AD therapies.60

Agents that inhibit the release of ET-1 or antagonize the ETA receptors on pericytes are worthy of consideration. In a murine model, C-type natriuretic peptide reversed the effects that were mediated by ET-1 and interrupted Aβ-provoked capillary constriction.105 Depletion of fibrinogen reduces CAA and cognitive decline in transgenic AD mice,110 and therefore, could represent an important therapeutic target.

Finally, because of their unique properties, in particular, their ability to withstand hypoxic conditions, perictyes should be considered as a viable option for cell-based therapies. In one study, mesenchymal stem cells that had differentiated into pericytes were stereotactically injected into the brains of mice genetically modified as an animal model for AD. The mice showed improved microcirculation and reduced levels of insoluble cortical and hippocampal Aβ,117 which suggested that pericyte implantation might provide a novel approach to the management of AD. Additional data indicate that pericytes harvested from temporal neocortex and cerebral ventricular zone proliferate readily in culture and are robust under storage.118,119

In summary, research into the NVU, in particular, the so-called “forgotten”120 cell—the pericyte—has yielded valuable insights into the pathogenesis of AD. With this knowledge, there is potential to develop promising avenues for treating what is perhaps the most relentless and refractory disease known to neuroscience.

Conclusions

Converging evidence has revealed that vascular pathology, rather than reflecting collateral or ancillary damage, is central to the pathophysiology of AD. Moreover, the markers for AD pathology that had been considered evidence of pure neurodegeneration, are mediators of vascular pathology. Pericytes, with their unique attributes as a locus for the interaction of multiple factors that contribute to neuronal integrity and stability, are perhaps positioned as a crucial nexus to resolve the pathophysiology of AD and establish a basis for the first effective disease-modifying interventions.

Abbreviations

Aβ: 

amyloid-beta

ACH: 

amyloid cascade hypothesis

AD: 

Alzheimer disease

AGE: 

advanced glycation end products

APOE: 

apolipoprotein E

APP: 

amyloid precursor protein

BBB: 

blood-brain barrier

CAA: 

cerebral amyloid angiopathy

CVD: 

cerebrovascular disease

ET-1: 

endothelin-1

IL: 

interleukin

LRP-1: 

low-density lipoprotein receptor-related protein-1

NOX: 

nicotinamide adenine dinucleotide phosphate oxidase

NVU: 

neurovascular unit

PDGF: 

platelet-derived growth factor

PDGFR-β: 

platelet-derived growth factor-beta

RAGE: 

receptor for advance glycation end products

ROS: 

reactive oxygen species

TNF: 

tumor necrosis factor

Declarations

Acknowledgement

None.

Funding

None.

Conflict of interest

The author has no conflicts of interest to declare.

Authors’ contributions

This manuscript is the sole work of SPC.

References

  1. Alzheimer A. Uber eine eigenartige Erkrankung der Hirnrinde. Allgemeine Zeitschrift fur Psychiatrie und Psychisch-gerichtliche Medizin 1907;64:146-148
  2. Alzheimer A, Stelzmann RA, Schnitzlein HN, Murtagh FR. An English translation of Alzheimer’s 1907 paper, “Uber eine eigenartige Erkrankung der Hirnrinde.”. Clin Anat 1995;8(6):429-431 View Article
  3. Davis KL, Thal LJ, Gamzu ER, Davis CS, Woolson RF, Gracon SI, et al. A double-blind, placebo-controlled multicenter study of tacrine for Alzheimer’s disease. The Tacrine Collaborative Study Group. N Engl J Med 1992;327(18):1253-1259 View Article
  4. Rogers SL, Friedhoff LT. The efficacy and safety of donepezil in patients with Alzheimer’s disease: Results of a US multicentre, randomized, double-blind, placebo-controlled trial. The Donepezil Study Group. Dementia 1996;7(6):293-303 View Article
  5. Reisberg B, Doody R, Stöffler A, Schmitt F, Ferris S, Möbius HJ, et al. Memantine in moderate-to-severe Alzheimer’s disease. N Engl J Med 2003;348(14):1333-1341 View Article
  6. Panza F, Lozupone M, Seripa D, Imbimbo BP. Amyloid-β immunotherapy for Alzheimer disease: Is it now a long shot?. Ann Neurol 2019;85(3):303-315 View Article
  7. Selkoe DJ. Alzheimer disease and aducanumab: adjusting our approach. Nat Rev Neurol 2019;15(7):365-366 View Article
  8. Blackard WG, Sood GK, Crowe DR, Fallon MB. Tacrine. A cause of fatal hepatotoxicity?. J Clin Gastroenterol 1998;26(1):57-59 View Article
  9. Orgogozo JM, Gilman S, Dartigues JF, Laurent B, Puel M, Kirby LC, et al. Subacute meningoencephalitis in a subset of patients with AD after Aβ42 immunization. Neurology 2003;61(1):46-54 View Article
  10. Kerchner GA, Boxer AL. Bapineuzumab. Expert Opin Biol Ther 2010;10(7):1121-1130 View Article
  11. Hachinski VC, Lassen NA, Marshall J. Multi-infarct dementia: A cause of mental deterioration in the elderly. Lancet 1974;2(7874):207-210 View Article
  12. Wallin A, Roman GC, Esiri M, Kettunen P, Svensson J, Paraskevas GP, et al. Update on Vascular Cognitive Impairment Associated with Subcortical Small-Vessel Disease. J Alzheimers Dis 2018;62(3):1417-1441 View Article
  13. Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW, et al. Gene dose of apolipoprotein E type4 allele and the risk of Alzheimer’s disease in late-onset families. Science 1993;261(5123):921-923 View Article
  14. Strittmatter WJ, Saunders AM, Schmechel D, Pericak-Vance M, Enghild J, Salvesen GS, et al. Apolipoprotein E: high-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. PNAS 1993;90(5):1977-1981 View Article
  15. Strittmatter WJ, Roses AD. Apolipoprotein E and Alzheimer’s disease. PNAS 1995;92(11):4725-4727 View Article
  16. Wisniewski T, Frangione B. Apolipoprotein E: A pathological chaperone protein in patients with cerebral and systemic amyloid. Neurosci Lett 1992;135(2):235-238 View Article
  17. Wisniewski T, Lalowski M, Golabek A, Vogel T, Frangione B. Is Alzheimer’s disease an apolipoprotein E amyloidosis?. Lancet 1995;345(8955):956-958 View Article
  18. Castellano JM, Kim J, Stewart FR, Jiang H, DeMattos RB, Patterson BW, et al. Human apoE isoforms differentially regulate brain amyloid-β peptide clearance. Sci Transl Med 2011;3(89):89ra57 View Article
  19. VIB-UAntwerp Center for Molecular Neurology. Alzheimer Disease and Frontotemporal Dementia Mutation Database (formerly Molgen Mutation Database) [Internet]. Available from: https://uantwerpen.vib.be/ADMutations. Accessed January 12, 2021
  20. Hof PR, Bouras C, Perl DP, Sparks DL, Mehta N, Morrison JH. Age-related distribution of neuropathologic changes in the cerebral cortex of patients with Down’s syndrome. Quantitative regional analysis and comparison with Alzheimer’s disease. Arch Neurol 1995;52(4):379-391 View Article
  21. De Strooper B. Proteases and proteolysis in Alzheimer disease: a multifactorial view on the disease process. Physiol Rev 2010;90(2):465-494 View Article
  22. Priller C, Bauer T, Mitteregger G, Krebs B, Kretzschmar HA, Herms J. Synapse formation and function is modulated by the amyloid precursor protein. J Neurosci 2006;26(27):7212-7221 View Article
  23. Potter H, Wisniewski T. Apolipoprotein E: Essential catalyst of the Alzheimer amyloid cascade. Int J Alzheimers Dis 2012;2012:489428 View Article
  24. Jucker M, Walker LC. Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature 2013;501(7465):45-51 View Article
  25. Hamaguchi T, Eisele YS, Varvel NH, Lamb BT, Walker LC, Jucker M. The presence of Aβ seeds, and not age per se, is critical to the initiation of Aβ deposition in the brain. Acta Neuropathol 2012;123(1):31-37 View Article
  26. Eisele YS. From soluble Aβ to progressive Aβ aggregation: Could prion-like templated misfolding play a role?. Brain Pathol 2013;23(3):333-341 View Article
  27. Deshpande A, Mina E, Glabe C, Busciglio J. Different conformations of amyloid beta induce neurotoxicity by distinct mechanisms in human cortical neurons. J Neurosci 2006;26(22):6011-6018 View Article
  28. Zurof L, Daley D, Black KL, Koronyo-Hamaoui M. Clearance of cerebral Aβ in Alzheimer’s disease: reassessing the role of microglia and monocytes. Cell Mol Life Sci 2017;74(12):2167-2201 View Article
  29. Qi XM, Ma JF. The role of amyloid beta clearance in cerebral amyloid angiopathy: more potential therapeutic targets. Transl Neurodegener 2017;6:22 View Article
  30. Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani SJ, Peske JD, et al. Structural and functional features of central nervous system lymphatic vessels. Nature 2015;523(7560):337-341 View Article
  31. Giannakopoulos P, Herrmann FR, Bussiere T, Bouras C, Kovari E, Perl DP, et al. Tangle and neuron numbers, but not amyloid load, predict cognitive status in Alzheimer’s disease. Neurology 2003;60(9):1495-1500 View Article
  32. Wright AL, Zinn R, Hohensinn B, Konen LM, Beynon SB, Tan RP, et al. Neuroinflammation and neuronal loss precede Aβ plaque deposition in the hAPP-J20 mouse model of Alzheimer’s disease. PLoS One 2013;8(4):e59586 View Article
  33. Cleveland DW, Hwo SY, Kirschner MW. Physical and chemical properties of purified tau factor and the role of tau in microtubule assembly. J Molec Biol 1977;116(2):227-247 View Article
  34. Morris M, Maeda S, Vossel K, Mucke L. The many faces of tau. Neuron 2011;70(3):410-426 View Article
  35. Alonso A, Zaidi T, Novak M, Grundke-Iqbal I, Iqbal K. Hyperphosphorylation induces self-assembly of tau into tangles of paired helical filaments/straight filaments. PNAS 2001;98(12):6923-6928 View Article
  36. Arriagada PV, Growdon JH, Hedley-Whyte ET, Hyman BT. Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer’s disease. Neurology 1992;42(3 Pt 1):631-639 View Article
  37. Choi SH, Kim YH, Hebisch M, Sliwinski C, Lee S, D’Avanzo C, et al. A three-dimensional human neural cell culture model of Alzheimer’s disease. Nature 2014;515(7526):274-278 View Article
  38. Wang L, Benzinger TL, Su Y, Christensen J, Friedrichsen K, Aldea P, et al. Evaluation of tau imaging in staging Alzheimer disease and revealing interactions between β-amyloid and tauopathy. JAMA Neurol 2016;73(9):1070-1077 View Article
  39. Hofman A, Ott A, Breteler MM, Bots ML, Slooter AJ, van Harskamp F, et al. Atherosclerosis, apolipoprotein E, and prevalence of dementia and Alzheimer’s disease in the Rotterdam Study. Lancet 1997;349(9046):151-154 View Article
  40. Maurer K, Volk S, Gerbaldo H. Auguste D and Alzheimer’s disease. Lancet 1997;349(9064):1546-1549 View Article
  41. Biessels GJ, Deary IJ, Ryan CM. Cognition and diabetes: a lifespan perspective. Lancet Neurol 2008;7(2):184-190 View Article
  42. Chui HC, Zarow C, Mack WJ, Ellis WG, Zheng L, Jagust WJ, et al. Cognitive impact of subcortical vascular and Alzheimer’s disease pathology. Ann Neurol 2006;60(6):677-687 View Article
  43. Kivipelto M, Helkala EL, Laakso MP, Hanninen T, Hallikainen M, Alhainen K, et al. Midlife vascular risk factors and Alzheimer’s disease in later life: longitudinal, population based study. BMJ 2001;322(7300):1447-1451 View Article
  44. Luchsinger JA, Reitz C, Honig LS, Tang MX, Shea S, Mayeux R. Aggregation of vascular risk factors and risk of incident Alzheimer disease. Neurology 2005;65(4):545-551 View Article
  45. Skoog I, Lernfelt B, Landahl S, Palmertz B, Andreasson LA, Nilsson L, et al. 15-year longitudinal study of blood pressure and dementia. Lancet 1996;347(9009):1141-1145 View Article
  46. Banks WA, Kastin AJ, Gutierrez EG. Interleukin-1 alpha in blood has direct access to cortical brain cells. Neurosci Lett 1993;163(1):41-44 View Article
  47. Heyman A, Fillenbaum GG, Welsh-Bohmer KA, Gearing M, Mirra SS, Mohs RC, et al. Cerebral infarcts in patients with autopsy-proven Alzheimer’s disease: CERAD, part XVIII. Consortium to Establish a Registry for Alzheimer’s Disease. Neurology 1998;51(1):159-162 View Article
  48. Viswanathan A, Rocca WA, Tzourio C. Vascular risk factors and dementia:How to move forward?. Neurology 2009;72(4):368-374 View Article
  49. Ellis RJ, Olichney JM, Thal LJ, Mirra SS, Morris JC, Beekly D, et al. Cerebral amyloid angiopathy in the brains of patients with Alzheimer’s disease: the CERAD experience, Part XV. Neurology 1996;46(6):1592-1596 View Article
  50. Hartz AM, Bauer B, Soldner EL, Wolf A, Boy S, Backhaus R, et al. Amyloid-β contributes to blood-brain barrier leakage in transgenic human amyloid precursor protein mice and in humans with cerebral amyloid angiopathy. Stroke 2012;43(2):514-523 View Article
  51. Greenberg SM, Charidimou A. Diagnosis of cerebral amyloid angiopathy: Evolution of the Boston criteria. Stroke 2018;49(2):491-497 View Article
  52. Viswanathan A, Greenberg SM. Cerebral amyloid angiopathy in the elderly. Ann Neurol 2011;70(6):871-880 View Article
  53. Sternberg EM. Neural regulation of innate immunity: A coordinated nonspecific host response to pathogens. Nat Rev Immunol 2006;6(4):318-328 View Article
  54. Meinl E, Krumbholz M, Hohlfeld R. B lineage cells in the inflammatory central nervous system environment: migration, maintenance, local antibody production, and therapeutic modulation. Ann Neurol 2006;59(6):880-892 View Article
  55. Norris JG, Benveniste EN. Interleukin-6 production by astrocytes: induction by the neurotransmitter norepinephrine. J Neuroimmunol 1993;45(1-2):137-145 View Article
  56. Gutierrez EG, Banks WA, Kastin AJ. Murine tumor necrosis factor alpha is transported from blood to brain in the mouse. J Neuroimmunol 1993;47(2):169-176 View Article
  57. Vukic V, Callaghan D, Walker D, Lue LF, Liu QY, Couraud PO, et al. Expression of inflammatory genes induced by beta-amyloid peptides in human brain endothelial cells and in Alzheimer’s brain is mediated by the JNK-AP1 signaling pathway. Neurobiol Dis 2009;34(1):95-106 View Article
  58. Sagare AP, Bell RD, Zlokovic BV. Neurovascular defects and faulty amyloid-β vascular clearance in Alzheimer’s disease. J Alzheimers Dis 2012;33(Suppl 1):S87-100 View Article
  59. Takuma K, Fang F, Zhang W, Yan S, Fukuzaki E, Du H, et al. RAGE-mediated signaling contributes to intraneuronal transport of amyloid-beta and neuronal dysfunction. PNASci 2009;106(47):20021-20026 View Article
  60. Kook SY, Hong HS, Moon M, Ha CM, Chang S, Inhee MJ. Aβ1-42-RAGE interaction disrupts tight junctions of the blood-brain barrier via Ca2+-calcineurin signaling. J Neurosci 2012;32(26):8845-8854 View Article
  61. Ellison MD, Krieg RJ, Povlishock JT. Differential central nervous responses following single and multiple recombinant interleukin-2 infusions. J Neuroimmunol 1990;28(3):249-260 View Article
  62. Montagne A, Barnes SR, Sweeney MD, Halliday MR, Sagare AP, Zhao Z, et al. Blood-brain barrier breakdown in the aging human hippocampus. Neuron 2015;85(2):296-302 View Article
  63. Grammas P, Ovase R. Inflammatory factors are elevated in brain microvessels in Alzheimer’s disease. Neurobiol Aging 2001;22(6):837-842 View Article
  64. Lendahl U, Nilsson P, Betsholtz C. Emerging links between cerebrovascular and neurodegenerative diseases-a special role for pericytes. EMBO Rep 2019;20(11):e48070 View Article
  65. Csiszar A, Ungvari Z, Edwards JG, Kaminski P, Wolin MS, Koller A, et al. Aging-induced phenotypic changes and oxidative stress impair coronary arteriolar function. Circ Res 2002;90(11):1159-1166 View Article
  66. Bell RD, Winkler EA, Singh I, Sagare AP, Deane R, Wu Z, et al. Apolipoprotein E controls cerebrovascular integrity via cyclophilin A. Nature 2012;485(7399):512-516 View Article
  67. Hughes S, Gardiner T, Hu P, Baxter L, Rosinova E, Chan-Ling T. Altered pericyte-endothelial relations in the rat retina during aging: implications for vessel stability. Neurobiol Aging 2006;27(12):1838-1847 View Article
  68. Kovacic JC, Moreno P, Nabel EG, Hachinski V, Fuster V. Cellular senescence, vascular disease, and aging: part 2 of a 2-part review: clinical vascular disease in the elderly. Circulation 2011;123(17):1900-1910 View Article
  69. Stewart PA, Magliocco M, Hayakawa K, Farrell CL, Del Maestro RF, Girvin J, et al. A quantitative analysis of blood-brain barrier ultrastructure in the aging human. Microvasc Res 1987;33(2):270-282 View Article
  70. Peppiatt CM, Howarth C, Mobbs P, Attwell D. Bidirectional control of CNS capillary diameter by pericytes. Nature 2006;443(7112):700-704 View Article
  71. Vates GE, Takano T, Zlokovic B, Nedergaard M. Pericyte constriction after stroke: the jury is still out. Nat Med 2010;16(9):959 View Article
  72. Bergers G, Song S. The role of pericytes in blood-vessel formation and maintenance. Neuro Oncol 2005;7(4):452-464 View Article
  73. Ozerdem U, Stallcup WB. Early contribution of pericytes to angiogenic sprouting and tube formation. Angiogenesis 2003;6(3):241-249 View Article
  74. Enge M, Bjarnegård M, Gerhardt H, Gustafsson E, Kalén M, Asker N, et al. Endothelium-specific platelet-derived growth factor-B ablation mimics diabetic retinopathy. EMBO J 2002;21(16):4307-4316 View Article
  75. Kale S, Hanai J, Chan B, Karihaloo A, Grotendorst G, Cantley L, et al. Microarray analysis of in vitro pericyte differentiation reveals an angiogenic program of gene expression. FASEB J 2005;19(2):270-271 View Article
  76. Takahashi H, Takata F, Matsumoto J, Machida T, Yamauchi A, Dohgu S, et al. Brain pericyte-derived soluble factors enhance insulin sensitivity in GT1-7 hypothalamic neurons. Biochem Biophys Res Commun 2015;457(4):532-537 View Article
  77. Armulik A, Abramsson A, Betsholtz C. Endothelial/pericyte interactions. Circul Res 2005;97(6):512-523 View Article
  78. Geraldes P, Hiraoka-Yamamoto J, Matsumoto M, Clermont A, Leitges M, Marette A, et al. Activation of PKC-delta and SHP-1 by hyperglycemia causes vascular cell apoptosis and diabetic retinopathy. Nat Med 2009;15(11):1298-1306 View Article
  79. Hammes HP, Lin J, Renner O, Shani M, Lundqvist A, Betsholtz C, et al. Pericytes and the pathogenesis of diabetic retinopathy. Diabetes 2002;51(10):3107-3112 View Article
  80. Bell RD, Winkler EA, Sagare AP, Singh I, LaRue B, Deane R, et al. Pericytes control key neurovascular functions and neuronal phenotype in the adult brain and during brain aging. Neuron 2010;68(3):409-427 View Article
  81. Baloyannis SJ, Baloyannis IS. The vascular factor in Alzheimer’s disease: a study in Golgi technique and electron microscopy. J Neurol Sci 2012;322(1-2):117-121 View Article
  82. Sagare AP, Sweeney MD, Makshanoff J, Zlokovic BV. Shedding of soluble platelet-derived growth factor receptor-β from human brain pericytes. Neurosci Lett 2015;607:97-101 View Article
  83. Yamagishi S, Yonekura H, Yamamoto Y, Fujimori H, Sakurai S, Tanaka N, et al. Vascular endothelial growth factor acts as a pericyte mitogen under hypoxic conditions. Lab Invest 1999;79(4):501-509
  84. Hyldahl RD, Xin L, Hubal MJ, Moeckel-Cole S, Chipkin S, Clarkson PM. Activation of nuclear factor-κB following muscle eccentric contractions in humans is localized primarily to skeletal muscle-residing pericytes. FASEB J 2011;25(9):2956-2966 View Article
  85. Scheibel AB, Fried I. Age-related changes in the pericapillary environment of the brain. Aging of the brain (Vol 22). Raven Press; 1983
  86. Zipser BD, Johanson CE, Gonzalez L, Berzin TM, Tavares R, Hulette CM, et al. Microvascular injury and blood-brain barrier leakage in Alzheimer’s disease. Neurobiol Aging 2007;28(7):977-986 View Article
  87. Mhatre M, Nguyen A, Kashani S, Pham T, Adesina A, Grammas P. Thrombin, a mediator of neurotoxicity and memory impairment. Neurobiol Aging 2004;25(6):783-793 View Article
  88. Montagne A, Nikolakopoulou AM, Zhao Z, Sagare AP, Si G, Lazic D, et al. Pericyte degeneration causes white matter dysfunction in the mouse central nervous system. Nat Med 2018;24:326-337 View Article
  89. Merlini M, Rafalski VA, Rios Coronado PE, Gill TM, Ellisman M, Muthukumar G, et al. Fibrinogen induces microglia-mediated spine elimination and cognitive impairment in an Alzheimer’s disease model. Neuron 2019;101(6):1099-1108 View Article
  90. Zlokovic BV. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat Rev Neurosci 2011;12(12):723-738 View Article
  91. Mussbacher M, Salzmann M, Brostjan C, Hoesel B, Schoergenhofer C, Datler H, et al. Cell type-specific roles of NF-κB linking inflammation and thrombosis. Front Immunol 2019;10:85 View Article
  92. Wu J, Hua Y, Keep RF, Schallert T, Hoff JT, Xi G. Oxidative brain injury from extravasated erythrocytes after intracerebral hemorrhage. Brain Res 2002;953(1-2):45-52 View Article
  93. Bell RD, Zlokovic BV. Neurovascular mechanisms and blood-brain barrier disorder in Alzheimer’s disease. Acta Neuropathol 2009;118(1):103-113 View Article
  94. Sengillo JD, Winkler EA, Walker CT, Sullivan JS, Johnson M, Zlokovic BV. Deficiency in mural vascular cells coincides with blood-brain barrier disruption in Alzheimer’s disease. Brain Pathol 2013;23(3):303-310 View Article
  95. Nation DA, Sweeney MD, Montagne A, Sagare AP, D’Orazio LM, Pachicano M, et al. Blood-brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nat Med 2019;25(2):270-276 View Article
  96. Zlokovic BV. Cerebrovascular effects of apolipoprotein E: implications for Alzheimer disease. JAMA Neurol 2013;70(4):440-444 View Article
  97. Halliday MR, Rege SV, Ma Q, Zhao Z, Miller CA, Winkler EA, et al. Accelerated pericyte degeneration and blood-brain barrier breakdown in apolipoprotein E4 carriers with Alzheimer’s disease. J Cereb Blood Flow Metab 2016;36(1):216-227 View Article
  98. Yamazaki Y, Shinohara M, Yamazaki A, Ren Y, Asmann YW, Kanekiyo T, et al. ApoE (apolipoprotein E) in brain pericytes regulates endothelial function in an isoform-dependent manner by modulating basement membrane components. Arterioscler Thromb Vasc Biol 2020;40(1):128-144 View Article
  99. Suo Z, Humphrey J, Kundtz A, Sethi F, Placzek A, Crawford F, et al. Soluble Alzheimers beta-amyloid constricts the cerebral vasculature in vivo. Neurosci Lett 1998;257(2):77-80 View Article
  100. Srikanth V, Maczurek A, Phan T, Steele M, Westcott B, Juskiw D, et al. Advanced glycation endproducts and their receptor RAGE in Alzheimer’s disease. Neurobiol Aging 2011;32(5):763-777 View Article
  101. Lue LF, Yan SD, Stern DM, Walker DG. Preventing activation of receptor for advanced glycation endproducts in Alzheimer’s disease. Curr Drug Targets CNS Neurol Disord 2005;4(3):249-266 View Article
  102. Sagare AP, Bell RD, Zhao Z, Ma Q, Winkler EA, Ramanathan A, et al. Pericyte loss influences Alzheimer-like neurodegeneration in mice. Nat Commun 2013;4:2932 View Article
  103. Wilhelmus MM, Otte-Höller I, van Triel JJ, Veerhuis R, Maat-Schieman ML, Bu G, et al. Lipoprotein receptor-related protein-1 mediates amyloid-beta-mediated cell death of cerebrovascular cells. Am J Pathol 2007;171(6):1989-1999 View Article
  104. Park L, Koizumi K, El Jamal S, Zhou P, Previti ML, Van Nostrand WE, et al. Age-dependent neurovascular dysfunction and damage in a mouse model of cerebral amyloid angiopathy. Stroke 2014;45(6):1815-1821 View Article
  105. Palmer JC, Barker R, Kehoe PG, Love S. Endothelin-1 is elevated in Alzheimer’s disease and upregulated by amyloid-β. J Alzheimers Dis 2012;29(4):853-861 View Article
  106. Belaidi E, Morand J, Gras E, Pepin JL, Godin-Ribuot D. Targeting the ROS-HIF-1-endothelin axis as a therapeutic approach for the treatment of obstructive sleep apnea-related cardiovascular complications. Pharmacol Ther 2016;168:1-11 View Article
  107. Nortley R, Korte N, Izquierdo P, Hirunpattarasilp C, Mishra A, Jaunmuktane J, et al. Amyloid β oligomers constrict human capillaries in Alzheimer’s disease via signaling to pericytes. Science 2019;365(6450):eaav9518 View Article
  108. Oshima K, Uchikado H, Dickson DW. Perivascular neuritic dystrophy associated with cerebral amyloid angiopathy in Alzheimer’s disease. Int J Clin Exp Pathol 2008;1(5):403-408
  109. Rensink AAM, Otte-Höller I, de Boer R, Bosch RR, ten Donkelaar HJ, de Waal RMW, et al. Insulin inhibits amyloid β-induced cell death in cultured human brain pericytes. Neurobiol Aging 2004;25(1):93-103 View Article
  110. Cortes-Canteli M, Paul J, Norris EH, Bronstein R, Ahn HJ, Zamolodchikov D, et al. Fibrinogen and beta-amyloid association alters thrombosis and fibrinolysis: a possible contributing factor to Alzheimer’s disease. Neuron 2010;66(5):695-709 View Article
  111. Mosconi L, Sorbi S, de Leon MJ, Li Y, Nacmias B, Myoung PS, et al. Hypometabolism exceeds atrophy in presymptomatic early-onset familial Alzheimer’s disease. J Nucl Med 2006;47(11):1778-1786
  112. Blair LJ, Frauen HD, Zhang B, Nordhues BA, Bijan S, Lin YC, et al. Tau depletion prevents progressive blood-brain barrier damage in a mouse model of tauopathy. Acta Neuropathol Commun 2015;3:8 View Article
  113. Kinney JW, Bemiller SM, Murtishaw AS, Leisgang AM, Salazar AM, Lamb BT. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement (N Y) 2018;4:575-590 View Article
  114. Fulga TA, Elson-Schwab I, Khurana V, Steinhilb ML, Spires TL, Hyman BT, et al. Abnormal bundling and accumulation of F-actin mediates tau-induced neuronal degeneration in vivo. Nat Cell Biol 2007;9(2):139-148 View Article
  115. Mills SM, Mallmann J, Santacruz AM, Fuqua A, Carril M, Aisen PS, et al. Preclinical trials in autosomal dominant AD: implementation of the DIAN-TU trial. Rev Neurol (Paris) 2013;169(10):737-743 View Article
  116. Wang WY, Tan MS, Yu JT, Tan L. Role of pro-inflammatory cytokines released from microglia in Alzheimer’s disease. Ann Transl Med 2015;3(10):136 View Article
  117. 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. Exp Neurol 2018;300:13-21 View Article
  118. Park TI, Feisst V, Brooks AE, Rustenhoven J, Monzo HJ, Feng SX, et al. Cultured pericytes from human brain show phenotypic and functional differences associated with differential CD90 expression. Sci Rep 2016;6:26587 View Article
  119. Paul G, Özen I, Christophersen NS, Reinbothe T, Bengzon J, Visse E, et al. The adult human brain harbors multipotent perivascular mesenchymal stem cells. PLoS One 2012;7(4):e35577 View Article
  120. Winkler EA, Sagare AP, Zlokovic BV. The pericyte: a forgotten cell type with important implications for Alzheimer’s disease?. Brain Pathol 2014;24(4):371-386 View Article
  • Exploratory Research and Hypothesis in Medicine
  • pISSN 2993-5113
  • eISSN 2472-0712
Back to Top

Pericytes and the Neurovascular Unit: The Critical Nexus of Alzheimer Disease Pathogenesis?

Steven P. Cercy
  • Reset Zoom
  • Download TIFF