Abstract
Alzheimer’s disease (AD) is the most prevalent form of dementia. Cerebrovascular dysfunction is one of the earliest events in the pathogenesis of AD, as well as in vascular and mixed dementias. Cerebral amyloid angiopathy (CAA), the deposition of amyloid around cerebral vessels, is observed in up to 90% of AD patients and in approximately 50% of elderly individuals over 80 years of age. CAA is a strong contributor to vascular dysfunction in AD. CAA-laden brain vessels are characterized by dysfunctional hemodynamics and leaky blood-brain barrier (BBB), contributing to clearance failure and further accumulation of amyloid-β (Aβ) in the cerebrovasculature and brain parenchyma. Mitochondrial dysfunction is increasingly recognized as an important early initiator of the pathogenesis of AD and CAA. The objective of this review is to discuss the effects of Aβ on cerebral microvascular cell function, focusing on its impact on endothelial mitochondria. After introducing CAA and its etiology and genetic risk factors, we describe the pathological relationship between cerebrovascular amyloidosis and brain microvascular endothelial cell dysfunction, critically analyzing its roles in disease progression, hypoperfusion, and BBB integrity. Then, we focus on discussing the effect of Aβ challenge on endothelial mitochondrial dysfunction pathways, and their contribution to the progression of neurovascular dysfunction in AD and dementia. Finally, we report potential pharmacological and non-pharmacological mitochondria-targeted therapeutic strategies which may help prevent or delay cerebrovascular failure.
Keywords: Alzheimer’s disease, amyloid, apoptosis, blood-brain barrier, cerebral amyloid angiopathy, endothelial cells, mitochondria, neurodegeneration, reactive nitrogen species, reactive oxygen species
INTRODUCTION
Dementia affects more than 40 million people in the world, and its incidence is expected to triple by 2050 [1]. Alzheimer’s disease (AD) is the most common form of dementia, although AD is known as a multifactorial disorder with multiple etiologies and different pathological manifestations. Mixed pathology dementias account for more than half of dementia cases, with amyloidosis and vascular disease being the most frequent combination [2–9]. The hallmark pathological characteristics of AD are senile or neuritic plaques of insoluble amyloid-β (Aβ) and neurofibrillary tangles (NFTs) of phosphorylated microtubule-associated protein tau (P-tau) [10]. In addition to these well-known lesions, pathological changes in the brain vasculature strongly contribute to clinical AD, as well as to vascular and mixed dementias [2, 11].
One of the most common but often overlooked pathological features of AD is cerebral amyloid angiopathy (CAA), the deposition of insoluble Aβ in cortical and leptomeningeal arteries, arterioles, and around the capillary walls [12, 13]. CAA is present in up to 90% of AD patients [14] and frequent in cognitively normal older individuals, at a prevalence of up to 50% after 80 years of age [15–17]. The perivascular accumulation of Aβ in CAA leads to cerebral endothelial cell (CEC) dysfunction and death [18]. Microvascular degeneration induced by CAA consequently results in hypoperfusion, neuroinflammation, blood-brain barrier (BBB) dysfunction, and oxidative stress, further contributing to the neurodegenerative process [18] and conferring substantial risk of vascular lesions in AD and other dementias [19]. This vascular viewpoint of the pathogenesis of AD and dementia is being investigated with increasing interest [2, 8, 20–22].
Cerebral endothelial cells control the chemical milieu of the brain by regulating molecular transport across the BBB and modulating cerebral blood flow (CBF). Vascular mechanisms of AD pathophysiology that are recognized among the Vascular Contributions to Cognitive Impairment and Dementia [8, 23] include chronic cerebral hypoperfusion, macro-and micro-hemorrhages, CEC dysfunction and death, capillary loss, neurovascular damage and impaired clearance [2, 8, 24–29]. The effects of altered cerebral hemodynamics on cognitive function have been extensively reviewed [11, 30–33], and small vessel damage and BBB breakdown are important mechanisms in AD pathogenesis that can contribute to the accumulation of Aβ and altered homeostasis in the brain [15, 34].
The objective of this review is to discuss the effects of Aβ on CEC dysfunction, specifically focusing on the pathways by which Aβ challenge affects the endothelial mitochondria. After introducing the clinical and neuropathological features, risk factors, and genetics of AD, this review describes the pathological effects of CAA on the cerebrovasculature such as hypoperfusion, ischemia, and BBB damage. Next, we describe the putative mechanisms responsible for amyloid-induced mitochondrial dysfunction in CECs, and analyze the specific components of such CEC mitochondrial dysfunction, encompassing energy metabolism, production of reactive oxygen (ROS), nitric oxide (NO) and reactive nitrogen species (RNS), calcium (Ca2+) crosstalk between the endoplasmic reticulum (ER) and mitochondria, and apoptosis. The review concludes with a discussion of therapeutic strategies against cerebral endothelial mitochondrial dysfunction and recommendations for future directions of relevant studies.
Alzheimer’s disease
Overview
AD is a progressive neurodegenerative disease in which gradual memory and cognitive impairments are typically preceded by extracellular deposition of Aβ as diffuse and insoluble plaques in the brain [35]. Aβ plaque deposition is followed by intracellular accumulations of NFTs and neurodegeneration [36]. Aβ is a product of β-secretase (BACE) and γ-secretase-catalyzed proteolysis of the amyloid-β protein precursor (AβPP), a single-pass transmembrane protein in the plasma membrane [10]. The Aβ plaques in AD brains are primarily comprised of Aβ42, but Aβ40 represents the main component of vascular amyloid deposits in CAA [12, 37]. Recent literature that utilized imaging and biomarker data from the large-scale database Alzheimer’s Disease Neuroimaging Initiative (ADNI) has reported that brain vascular dysfunction precedes both Aβ and tau pathology, and cognitive impairment in AD [38]. Preclinical studies also argue for the importance of vascular, rather than brain parenchymal amyloid, in inducing early cognitive impairment in mice [11, 22, 39, 40].
Risk factors and genetics
Aging is the most common and widely known risk factor for developing AD [41]. Other risk factors for AD include the female sex for increased risk of onset but male sex for duration of survival after clinical diagnosis [42], low educational and occupational status, low mental status, low mental and physical activity [43], and cardiovascular risk factors such as hypercholesterolemia, hypertension, cerebrovascular diseases [44], hyperhomocysteinemia [45], smoking, obesity [46], and diabetes mellitus [47]. Traumatic brain injury (TBI) has also recently been recognized as a possible risk factor for AD [48] and associated with cerebrovascular dysfunction [49], but the exact link between TBI and AD still remains under debate [50].
The ε4 allele of the apolipoprotein E (APOE) gene is the most common genetic risk factor [51] for AD. Sporadic AD, or late-onset AD, which occurs in older patients who are over the age of 65 years [52], accounts for 95–99% of all AD cases [52, 53]. Familial AD is referred to as early-onset AD, occurring in patients younger than 65 years of age [52]. Familial AD involves mutations in the presenilin-1 (PS1), presenilin-2 (PS2), and APP genes [54]. Main mutations on Aβ which lead to familial AD include A2V (A2V), English (H6R), Tottori (D7N), K16N (K16N), Flemish (A21G), Osaka (E22Δ), Italian (E22K), Dutch (E22Q), Arctic (E22G), Iowa (D23N), Piedmont (L34V), Swedish (KM670/671NL), and Indiana (V717F) [55–57]. Most familial forms of AD are marked by increased kinetics of Aβ aggregation [55], which results in early age of onset. Multiple mutations in the Aβ sequence and other aggregation-prone proteins such as ABri and Adan are particularly associated to early-onset aggressive forms of CAA and related to dementia with cerebral hemorrhage [18, 58].
Cerebral amyloid angiopathy
In addition to the extracellular accumulation of Aβ plaques from frontal and temporal lobes and the hippocampus and limbic systems to other regions of the brain parenchyma, a hallmark amyloid pathology of AD is the deposition of Aβ in the walls of cortical and leptomeningeal arteries, arterioles and capillaries, termed CAA [12]. Emerging evidence suggests that there are two types of CAA: arterial and capillary/pericapillary CAA. In arterial CAA, insoluble Aβ deposits in the vessels’ tunica media and adventitia along the perivascular space [12, 59, 60]. Pericapillary CAA is suggested to be marked by insoluble Aβ aggregates along the glia limitans surrounding the capillary and may indicate the initial stages of Aβ accumulation due to impaired glymphatic clearance of Aβ and other waste products [13]. Capillary CAA is the deposition of Aβ within the capillary wall (i.e., between the endothelium and basement membrane) [13].
CAA is one of the most common cerebral small vessel diseases and the most frequent cause of lobar intracerebral macro- and micro-hemorrhages in elderly individuals [15, 37, 61]. CAA is also a major risk factor for white matter hyperintensities observed via magnetic resonance imaging (MRI), hypoperfusion and ischemia, BBB damage in cerebral arterioles and capillaries, and molecular dysfunctions of CECs [37]. Therefore, CAA confers substantial vascular contribution to the pathogenesis of AD [8, 19]. Hereditary CAA is most commonly caused by Dutch, Italian, Arctic, Iowa, Flemish, and Piedmont mutations in the APP gene, as well as the Icelandic mutation in the CST3 gene (L68Q), and the British and Danish mutations in the ITM2B gene [62]. CAA has been recognized as a cause for intracerebral hemorrhage, brain hypoperfusion, ischemia and BBB dysfunction, and all forms of cerebrovascular disease discussed below [22, 63].
Hypoperfusion and ischemia
A prolonged reduction of CBF, or hypoperfusion, due to hypotension, cardiovascular disease, or CAA, limits glucose and oxygen delivery and impairs brain function. An increasing amount of evidence has demonstrated that cerebrovascular dysfunction may precede and exacerbate amyloid accumulation, linking cardiovascular risk factors to the development of CAA and AD [64, 65]. Hypoperfusion seems to accelerate the development of CAA, which, in turn, is associated with the development of cortical microinfarctions [66] and reduced CBF, establishing a degenerative feed-forward process for the brain’s neurovascular unit. Indeed, cerebrovascular hypoperfusion could be one of the underlying inducers for the development of AD, as drugs that treat hypoperfusion are able to alleviate some of the AD symptoms [67]. However, the cause-effect relationship between CAA and hypoperfusion is still unclear [68]. It appears that hypoperfusion could result in vascular damage that leads to Aβ accumulation which, in turn, leads to more vascular damage. For example, accumulating evidence supports the role of Aβ in mediating vasoconstriction through the reduction of NO bioavailability as a direct consequence of increased ROS levels (discussed in the NO section below).
In support of the concept that cerebral hypoperfusion precedes amyloid accumulation, in the APP Swedish and Indiana mutation mice (APP-SweInd), cerebral hypoperfusion was shown to accelerate Aβ deposition [69]. Actually, cerebral hypoperfusion alone resulted in a decrease in learning and memory, and increased Aβ40 and Aβ42 levels in the hippocampus [70]. Similar results were obtained in transgenic Swedish, Dutch and Iowa, mutation (Tg-SwDI) mice, where cerebral hypoperfusion increased Aβ40 and Aβ42 in the cerebral vasculature and increased microinfarctions [71]. It was further demonstrated that APP/PS1 mice with carotid occlusion shifted Aβ toward high molecular weight oligomeric species [72], suggesting an increase in amyloid burden and disease progression. Additionally, hypertension accelerated the development of cognitive deficits, Aβ deposition in the microvasculature, and BBB dysfunction in the Tg-SwDI mice [65].
The increase in Aβ40/Aβ42 levels may be due to increased activity of the amyloidogenic pathway through the upregulation of BACE and γ-secretase [70]. Hypoxia inducible factor (HIF), a transcription factor that is activated due to hypoxia, was shown to induce BACE-1 expression [73], therefore possibly contributing to the hypoperfusion-induced Aβ accumulation. In an in vitro model of cerebral hypoperfusion using oxygen and glucose deprivation, brain capillary endothelial cells upregulated the expression of HIF-1, BACE1, AβPP, and Aβ42 levels [74]. However, other studies have portrayed a possible neuroprotective role of HIF [75, 76]. It is possible that the activation of HIF-1 could mediate protective mechanisms during acute hypoxic conditions to prevent cell death but that its prolonged activation could lead to BACE-1 activation, and eventually cell death [77].
The blood-brain barrier in CAA and AD
The BBB is formed by tight junction proteins that connect CECs, which are surrounded by the basement membrane and astrocytic end feet, thereby creating a selectively permeable barrier [78]. Cerebral endothelial cells strictly regulate the transport of solutes between the lumen of blood vessels and the interstitium of the brain parenchyma, and, consequently, regulate brain homeostasis [79]. As a result, the BBB is necessary to prevent the entry of toxins, waste products, or other detrimental agents into the brain. Dysfunction of CECs can result in a disrupted, or “leaky,” BBB, which allows the influx of toxic and undesirable molecules into the brain, impairing brain homeostasis, and inducing inflammation and aberrant neurovascular function. It is hypothesized that BBB dysfunction leads to neuronal damage and Aβ accumulation in the brain [79]. This may occur progressively throughout the neurovascular unit, comprised of CECs, pericytes, smooth muscle cells, astrocytes, microglia, oligodendroglia, and neurons [80]. For example, hypoperfusion and Aβ accumulation can lead to P-tau and NFT accumulation and injury in neurons and activate microglia [80].
Substantial loss of endothelium is observed in human AD brains [81]. Along with decreased mitochondrial content in CECs [82], brain biopsies [82], and MRI of AD patients [83] have shown BBB dysfunction and leakage. Transgenic murine models of CAA and AD have further corroborated such BBB dysfunction [84]. Furthermore, in humans with AD pathology (CSF Aβ42 positive), cerebrovascular capillary and BBB damage was evident. Interestingly, they also reported that BBB damage was present in humans with early cognitive dysfunction (CSF Aβ42 negative), demonstrating that BBB damage is present before Aβ accumulation and at the onset of mild cognitive dysfunction, therefore making it capable to serve as a predictor of cognitive function [21]. These data suggest that BBB dysfunction may be an early mediator of AD pathology.
BBB leakage or dysfunction is primarily mediated by the loss of tight junction (TJ) proteins. Cerebrovascular endothelial TJs include integral membrane proteins such as occludin, claudins, and junctional adhesion molecules (JAMs). These, in turn, interact with intracellular scaffold proteins such as zonula occludens and cytoskeletal proteins. Surprisingly, only claudins, and not occludin and JAMs, appear to be essential in BBB integrity (reviewed in [85]). Given the importance of BBB in brain homeostasis, it is of no surprise that BBB dysfunction observed in CAA and AD pathology may be mediated by the loss of TJs. An increase in BBB permeability coupled to a reduction of TJ proteins claudin-1 and claudin-5 was observed in isolated rat brain microvessels treated with human Aβ40 and in microvessels from transgenic mice expressing hAPP with the Swedish mutation (Tg2576) [86]. Furthermore, claudin-5 expression, and to a lesser extent occludin, were downregulated in multiple areas of human postmortem AD brains [87]. Interestingly, the authors also demonstrated that there is an inverse correlation between cortical claudin-5 and occludin expression, and insoluble Aβ40 and CAA. Overall, BBB dysfunction contributes to the development of CAA and AD pathology through the downregulation of TJ proteins, where claudin-5 appears to be the main player.
Energy metabolism of endothelial mitochondria
As mitochondria represent the energy source for our brain cells, mitochondrial damage is likely to be one of the earliest events in the development of AD [88–95]. Mitochondrial dysfunction can not only induce energetic failure in the AD brain, but also contribute to the activation of executioner caspases, such as caspase 3, thereby mediating neuronal, vascular and glial cell death, and the progression of AD pathology [96–100].
Physiological metabolism
Compared to peripheral endothelial cells, CECs are much more dependent on mitochondrial respiration for their energy metabolism and survival. Mitochondrial volume in rat brain capillary CECs was 8–11% of the total cytoplasmic volume, or 2–4 times the mitochondrial volume of peripheral endothelial cells [101]. Their higher susceptibility to apoptosis and disruptions in TJs due to hypoxic or ischemic conditions also suggest their dependence on mitochondrial metabolism [102]. Glucose is the main obligatory metabolic substrate of the human adult brain and its CECs [103]. A 55-kDa isoform of glucose transporter 1 (GLUT1) imports glucose into the CECs [104]. Glucose then undergoes glycolysis, and subsequently lactate fermentation or mitochondrial metabolism, or the pentose-phosphate pathway [103]. The adenosine triphosphate (ATP) turnover rate and the percentage of ATP produced by the mitochondria relative to overall CEC metabolism are not yet clear.
Effects of Aβ on mitochondrial metabolism
In CECs, it is not yet known the mechanisms by which Aβ disrupts mitochondrial metabolic pathways and systems such as the tricarboxylic acid (TCA) cycle, electron transport chain (ETC), and oxidative phosphorylation, nor the enzymes aberrantly affected by Aβ. However, due to the conserved structure-function of enzymes in many of these metabolic pathways, it is possible that Aβ may affect these metabolic processes and ATP production in CECs in similar ways as in non-synaptic brain, neuronal, and astrocytic mitochondria.
Exposure to Aβ25–35, which is the shortest fragment capable of forming β-sheet fibrils and retaining the toxicity of the full length Aβ40/42 peptides [105], led to decreased ATP production by 42% in neurons and by 28% in astrocytes compared to respective control cells [106]. In non-synaptic brain mitochondria, 30-min exposure to Aβ25–35 inhibited pyruvate dehydrogenase and α-ketoglutarate dehydrogenase activity, consequently downregulating pyruvate production and TCA cycle activity [107]. In cerebellar granule neurons, exposure to Aβ42 impaired ETCs nicotinamide adenine dinucleotide (NADH)-ubiquinone oxidoreductase (Complex I) and cytochrome c (CytC) oxidase (Complex IV) activities [108] and bound to Complex IV in human neuroblastoma cells [109]. Another metabolic damage caused by amyloid proteins is the impairment of oxidative phosphorylation. Both AβPP and Aβ40 bound to subunit α of F1F0-ATP synthase and partially inhibited ATP production in neuroblastoma cells [110]. However, immortalized microvascular CECs challenged with the aggressive vasculotropic variant Aβ40-E22Q, did not show a decrease in mitochondrial ATP production [111].
In this study, amyloid peptides exerted differential effects on SH-SY5Y (neuronal) and CECs, inducing a slight decrease in ATP levels in neuronal cells and a substantial increase in CECs. A possible explanation is that these cell types handle mitochondrial dysfunction by differentially resorting to aerobic glycolysis [112]. Additional studies in primary CECs are needed to clarify whether this is a characteristic linked to the immortalized nature of the human CEC used (hCMEC/D3), or if different pathways characterize the neuronal/astrocytic versus endothelial mitochondrial metabolic effects of Aβ.
ENDOTHELIAL PRODUCTION OF REACTIVE OXYGEN SPECIES
Metabolism and ROS production in the mitochondria are interconnected. In the ETC, Complex I and coenzyme Q (CoQ)-CytC reductase (Complex III) are primarily responsible for producing superoxide anions (O2−) [113]. Complex I produces O2− in the process of oxidizing NADH into NAD and pumping a proton from the mitochondrial matrix into the intermembrane space [113]. Complex III produces O2− as it oxidizes CoQ to reduce CytC and pump a proton into the intermembrane space [113]. Molecules of O2− are potent oxidants and, therefore, need to be reduced and “detoxified.” In the mitochondrial matrix, superoxide dismutase (SOD) 2 reduces O2− into H2O2, a less toxic ROS [114]. Then, glutathione peroxidase (GPX) catalyzes the reduction of H2O2 into H2O through the oxidation of reduced glutathione (GSH) into its oxidized form (GSSG) [114]. Catalase in the mitochondrial matrix can also convert H2O2 into water and molecular oxygen [114]. Glutathione biosynthesis is catalyzed by glutathione reductase using the oxidation of reduced nicotinamide adenine dinucleotide phosphate (NADPH) into NADP and is crucial in antioxidant activities in the mitochondria [114]. In the intermembrane space, copper and zinc-containing SOD1 reduces O2− into H2O2, and GPX reduces H2O2 into H2O [114].
Damage to the ETC and oxidative phosphorylation due to Aβ in CEC mitochondria can lead to excessive production of mitochondrial ROS, oxidative stress, further damage to the mitochondria and cell, and exacerbation of vascular amyloid pathology. Changes in mitochondria-specific redox homeostasis due to Aβ have been investigated in transgenic CAA mouse models [115, 116]. Proteomic analysis of CECs isolated from cerebral arteries from the Circle of Willis and its main branches in the double transgenic APP-SweInd mice showed upregulated expressions of mitochondrial copper-zinc SOD1 and CytC oxidase subunit 6C [115]. A similar double transgenic CAA mouse model of Tg2576 that also expresses PS1 (L166P) showed that there was upregulated SOD2 expression and O2− production in CECs of leptomeningeal arteries [116]. The mitochondria-derived ROS, in turn, exacerbated CAA pathology in such mice [116], suggesting a downward spiral of Aβ accumulation and oxidative stress in the cerebrovasculature.
Recent studies by our group also clearly demonstrated an increase in H2O2 production by mitochondria isolated after treatment of human CEC with Aβ peptides [111], accompanied by the loss of mitochondrial membrane potential (Δψ) [111, 117], both prevented in presence of carbonic anhydrase inhibitors (CAIs). However, the mechanism responsible for the increase in H2O2 induced by Aβ is not fully understood. While other studies have investigated the effect of exogenous Aβ on the oxidative stress and ROS production of CECs, not necessarily specifically to the mitochondria, it is likely that the redox responses in CECs include mitochondrial ROS production and antioxidant enzymes. In bEnd3 mouse CEC line and primary rat CECs, Aβ25–35 exposure led to increased cellular ROS production and downregulated cellular SOD activity [118, 119], as well as increased cellular lipid peroxidation, decreased GSH/GSSG, and downregulated cellular catalase, glutathione-S-transferase, and GPX activities [119]. Interestingly, exposure to lower concentrations of Aβ may allow CECs to recover from mitochondrial and overall cellular oxidative stress. Rat CECs (RBE4) exposed to low dosage of Aβ40 had increased cellular ROS levels and decreased GSH/GSSG ratio at 3, 6, and 12 hours, which normalized by 24 hours [120]. Glutathione reductase expression also increased at 12 and 24 hours, which also suggests an antioxidant recovery effect in CECs [120].
NITRIC OXIDE AND REACTIVE NITROGEN SPECIES
The vasoactive substance NO is essential for the regulation of CBF and maintenance of cerebral function. Proper regulation of vascular tone and CBF is essential to couple brain metabolic demand with supply. Nitric oxide is synthesized by the nitric oxide synthases (NOS) and promotes vascular relaxation through its actions on vascular smooth muscle cells. There are three main NOS isoforms: neuronal NOS (nNOS) and endothelial NOS (eNOS), which are activated by Ca2+-calmodulin, and Ca2+-independent inflammation-induced inducible NOS (iNOS). Various studies have proposed the existence of a mitochondrial localized NOS (mtNOS) [121, 122], but these findings have been challenged (discussed in detail in [123, 124]).
Given the importance of CBF in the development and progression of CAA and neurodegeneration [125], the role of NOS in the cerebrovasculature has been widely studied. Aβ1–40 has been shown to induce vasoconstriction in cerebral vascular endothelial cells in vitro and in vivo [126]. The expression of capillary eNOS was found to negatively correlate with the amount of NFTs and senile plaques in postmortem human brains [127]. In addition, mice lacking eNOS (eNOS−/−) displayed increased levels of AβPP and BACE1 in whole brain tissue [128]. Indeed, partial deficiency of eNOS (eNOS−/+) is enough to induce vascular changes that result in CAA, BBB breakdown, and cognitive deficits [129]. Therefore, a growing number of studies demonstrate a crucial role of eNOS for proper endothelial function and a potential link between decreased eNOS expression and activity and the development of CAA.
Interestingly, PC12 cells with the double Swedish mutation (APPsw; K670M/N671L) have increased NO levels possibly due to increased NOS activity [130]. This increase in NO levels may be mediated by iNOS, which was found to be elevated in AD postmortem brains [127]. Although enhanced NO production seems to be a beneficial, rather than detrimental effect, NO availability is severely impaired in the presence of ROS, specifically O2−, due to the spontaneous formation of peroxynitrite (ONOO−), which has oxidizing and nitrating capabilities. This is further supported by data demonstrating that administration of a ROS scavenger to Tg-SwDI mice enhanced vascular function [40], indicating that ROS may decrease NO bioavailability, thus contributing to vascular dysfunction and vasoconstriction in CAA.
The spontaneous formation of ONOO− occurs faster than the O2−dismutation by MnSOD [131], suggesting that even during normal MnSOD activity and expression, the formation of ONOO− is evident. Most importantly, NO inhibits SOD by preventing O2− binding, therefore increasing intracellular ROS and possibly RNS [132]. Interestingly, phosphorylation of eNOS at serine-1177 increases O2− promotion [133], an event that is observed in the brain of the Tg2576 mouse model of amyloidosis [134]. Furthermore, nitrotyrosine content, an indicator of ONOO− damage, was increased in brains from AD patients when compared to controls [135, 136]. Importantly, in Tg2576 mice, an increase in 3-nitrotyrosine levels and amyloid deposition were observed in cerebrovascular endothelial cells [137]. In cultured cortical neurons, treatment with a NOS inhibitor was able to reduce reactive species after Aβ22–35 treatment suggesting that ONOO− is the major reactive species formed in these conditions [138]. The excessive ONOO− formation severely impairs NO bioavailability leading to vasoconstriction and reduced CBF. In a mouse model of AD, endothelial-NO bioavailability was severely impaired [139], which could be secondary to excessive ONOO− formation. This data is further supported by studies in human umbilical vein endothelial cells where Aβ25–35 decreased NO levels [140].
In ratCECs, mitochondrial membrane depolarization induced NO production through a Ca2+-mediated activation of eNOS [141]. Elevated NO inhibits cellular respiration in vascular endothelial cells by inhibiting Complex IV [142, 143] and is associated with mitochondrial dysfunction in PC12 cells with the AβPPsw K670M/N671L mutation [130]. Membrane depolarization, with elevated ROS production, in particular O2−, will then induce production of ONOO−, depleting the NO pool. Therefore, it appears that Aβ deposition in the cerebrovasculature is accompanied by an increase in ONOO− damage and consequently a reduction of NO bioavailability resulting in vasoconstriction and reduction of CBF.
The formation of nitrotyrosine has also been observed to influence Aβ aggregation. Nitrosylation of Aβ peptide induced formation of Aβ oligomers [144] which are known to be the most toxic aggregation species [145] for multiple brain cell types including CECs and smooth muscle cells [146–148]. Other groups have reported that nitrotyrosine promotes Aβ aggregation and plaque formation [149]. Although the data available is still controversial, it suggests a possible role of RNS, most specifically ONOO−, in the regulation of Aβ toxicity and plaque formation. On the other hand, coadministration of ONOO− with Aβ in rat CECs, did not exacerbate vascular cytotoxicity [150]. Overall, the data suggests that NO and RNS could potentiate endothelial mitochondrial dysfunction in AD. This can not only cause cerebrovascular dysfunction due to the absence of NO, but also DNA damage due to the reactive properties of ONOO− [151].
CALCIUM, ENDOPLASMIC RETICULUM, AND MITOCHONDRIAL PERMEABILITY TRANSITION PORE
The endoplasmic reticulum (ER) is primarily involved in the regulation of Ca2+ homeostasis. Under physiological conditions, the mitochondria can uptake Ca2+ after it has been released from the ER to increase metabolic activity through the activation of several enzymes [152]. In pathological conditions leading to sustained activation of ER stress, caspase activation and CytC release is activated through the mitochondria [153]. Although these mechanisms have been extensively studied in neuronal cells [154, 155], evidence suggest that similar mechanisms may be involved in Aβ-induced brain endothelial dysfunction.
Rat brain endothelial cells treated with Aβ40, the primary Aβ isoform in CAA, displayed increased ER stress response markers and disrupted Ca2+ homeostasis as observed by a decrease in ER Ca2+ and an increase in cytosolic and mitochondrial Ca2+ [120, 156]. Both, Aβ and ER Ca2+ depletion induce the formation of a transient receptor potential (TRP) channel in the plasma membrane provoking an increase in Ca2+ conductance, which further leads to an increase in BBB permeability and endothelial dysfunction [157, 158]. However, whether the effects of Aβ on ER Ca2+ depletion are mediated through the TRP channel or through other mechanisms is still unclear.
The mitochondria are key mediators of cell death. Under conditions of high matrix Ca2+, elevated ROS production, and loss of Δψ, mitochondrial permeability transition (mPT) can be induced. Pathological mPT leads to the formation of an irreversible, non-selective pore termed the mPT pore (mPTP) resulting in mitochondrial swelling, outer membrane rupture, and the induction of cell death. ER calcium depletion, loss of Δψ, and CytC release observed in Aβ40 treated rat, murine and bovine endothelial cells [156, 159] may be indicative of mPTP formation. Further supporting this data, Yin and collaborators demonstrated that inhibition of the mPTP using Cyclosporin A (CsA) was able to prevent Aβ-induced release of the mitochondrial apoptotic protein second mitochondria-derived activator of caspases (Smac) [105]. In primary cultures of rat astrocytes and hippocampal neurons, a reduction of mitochondrial inorganic phosphate prevented Aβ-induced mitochondrial depolarization and cell death, possibly through inhibition of Ca2+-induced mPTP [160]. However, in human CECs treated with the aggressive vasculotropic mutant Aβ40-E22Q, both mitochondrial and cytoplasmic Ca2+, measured by Fluo-4 and Rhod-2 fluorescence, respectively, were significantly decreased [111]. Taken together, the data suggests that Aβ may disrupt Ca2+ homeostasis, leading to an increase in BBB permeability and mitochondria-mediated cell death through the release of pro-apoptotic factors, although further research is needed to clarify the underlying mechanisms.
MITOCHONDRIA-MEDIATED APOPTOSIS IN CEREBRAL ENDOTHELIAL CELLS
Physiology of intrinsic apoptosis
Intrinsic apoptosis can be activated by aberrant metabolic activity in pathways such as oxidative phosphorylation [161], overproduction of ROS and RNS [161], DNA damage [162], ER stress [163], and ER release of excess Ca2+ into the mitochondria [163]. Aberrant intracellular cascade signaling and DNA-binding proteins can also induce intrinsic apoptosis by inhibiting anti-apoptotic kinases that promote cell growth such as protein kinase B (Akt) [164] or activating pro-apoptotic kinases such as apoptosis signal-regulating kinase 1 (ASK1) [165, 166] and c-Jun N-terminal kinase [167].
These intracellular triggers activate Bcl-2-associated X (Bax) and Bcl-2 homologous antagonist killer (Bak) channels [168], which allow the translocation of CytC released from oxidized cardiolipin (also released by mPTP) [169, 170] and the pro-apoptotic signaling protein Smac, also known as direct inhibitor of apoptosis protein (IAP) binding protein with low pI homolog (Diablo) [168]. Translocated CytC joins apoptotic protease activating factor 1 and procaspase-9 to form the apoptosome [171]. The activated apoptosome activates caspase-3 and −7, which subsequently catalyze substrate proteolysis, and ultimately cell death [171].
Aβ-induced cerebral endothelial intrinsic apoptosis
Aβ has been shown to directly activate mitochondria-mediated apoptotic pathways as well as key signal transduction kinases that have downstream pro-apoptotic mitochondrial targets in CECs [146]. Bovine brain microvascular endothelial cells can uptake human Aβ40, likely through ATP-dependent endocytosis [147, 172], suggesting that internalized Aβ40 may induce intrinsic apoptosis through ER stress, ROS/RNS production, and intracellular Ca2+ overload.
This concept is supported by the Aβ-induced early ER stress in RBE4 cells, shown by increased expressions of cyclic AMP-dependent transcription factor 4 (ATF4) and 6a (ATF6a) [156], Grp78, and xbp-1 [156, 173], and consequent early and prolonged depletion of ER Ca2+ and increase in cytosolic Ca2+ [120, 156]. These events were followed by upregulation of caspase-9, caspase-3, CytC release, and loss of Δψ [156]. Alongside ER stress, the intracellular accumulation of ROS such as O2− and toxic molecules such as 4-hydroxynoneal caused by Aβ40 exposure may lead to intrinsic apoptosis, measured by loss of Δψ [174]. Another indication of intrinsic apoptosis is damage to the mitochondrial DNA, measured by reductions in long-chain PCR products. Aβ25–35 was shown to induce mitochondrial DNA damage after 24 and 48 hours in murine CECs [159], which was accompanied by CytC release, caspase-3 and −8 activation. Accordingly, vasculotropic Aβ40 mutants (such as the Dutch, Piedmont, and Iowa mutants), as well as wild-type (WT)-Aβ40, all triggered CytC release and caspase activation pathways [117, 146, 147]. Mitochondrial apoptotic pathways may also be engaged as a secondary step during extrinsic apoptotic cascades. Importantly, the pro-apoptotic effects induced by Aβ peptides in cerebrovascular cells, as well as in neuronal and glial cells, have been mainly attributed to oligomers and protofibrils [117, 146, 147, 175, 176], which are typically accumulated extracellularly and acting as alternative receptor ligands.
Physiology of extrinsic apoptosis
Extracellular death ligands, such as FasL and tumor necrosis factor (TNF) α, bind to the extracellular domains of transmembrane death receptors (DRs) to trigger extrinsic apoptosis [177]. Common death receptors include the cluster of differentiation 95 (CD95 or Fas), TNF receptors (TNF-R), and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) DRs [178–180]. The ligand-DR binding allows the DR’s cytosolic domain to recruit adaptor proteins such as the Fas-associated death domain protein (FADD) [179]. These adaptor proteins then recruit multiple procaspase-8 molecules, which then undergo auto-cleavage and activation [177]. Caspase-8 cleaves and activates BH3 interacting-domain death agonist (Bid) into truncated Bid (tBid), which activates the Bax/Bak proteins, while also cleaving procaspases-3 and −7 into their active caspase forms for substrate proteolysis and cell death [171, 180].
Effect of Aβ on extrinsic endothelial apoptosis
These pathways have been investigated in Aβ-exposed endothelial cells using primary murine, rat, and human CECs, mouse (bEnd.3), rat (REB4), and human (hCMEC/D3) brain endothelial cell lines, and CECs derived from AβPPsw mice. In hCMEC/D3 cells, Aβ40-E22Q oligomers have been shown to directly bind to TRAIL DR4 and DR5, leading to increased caspase-8 activity, downregulation of cellular FADD-like IL-1β-converting enzyme-inhibitory protein (c-FLIP, an inhibitor of caspase-8), consequent Bid cleavage and CytC release [148]. Translocation of Bax from the cytosol to mitochondria has also been previously shown [181] in human CECs. Interestingly, Aβ may also trigger an extrinsic-like apoptotic mechanism through the activation of CEC surface receptors other than apoptotic death receptors, such as the receptor for advanced glycation endproducts (RAGE) [182] and scavenger receptor cluster of differentiation 36 (CD36) [183].
A transmembrane multi-ligand receptor that’s part of the immunoglobulin (Ig) superfamily, RAGE’s physiological function involves inflammatory and immune responses [184]. Previous studies have shown that RAGE directly binds to Aβ40 and Aβ42 [185] and may be involved in their apical-to-basolateral transport in CECs [186]. In bEnd.3 cells, Aβ1–42 (WT) activated RAGE and subsequently induced ER stress, marked by ER stress markers 78-kDa glucose-regulated protein (Grp78), X-box binding protein 1 (xbp-1), and CCAAT-enhancer-binding protein homologous protein [182]. This ER stress subsequently led to both mitochondria-dependent and -independent apoptosis. Mitochondria-dependent apoptosis was shown through an increased ratio of Bax/Bcl-2 expression, and mitochondria-independent apoptosis was seen by an increased expression of caspase-12. Adiponectin has been recently shown to decrease RAGE expression and attenuate apoptosis in Aβ-challenged bEnd3 cells. However, binding of Aβ oligomers to RAGE has been recently debated [187].
CD36 is a multi-ligand, class B transmembrane glycoprotein receptor that is involved in fatty acid transport [188] and negative regulation of angiogenesis in microvascular endothelial cells [189]. In Tg2576 mice, the binding of Aβ40 to CD36 was found necessary for NADPH oxidase-mediated ROS production and oxidative stress in cerebral endothelial cells of penetrating arterioles [183].
Signal transduction kinases
In addition to the extrinsic, extrinsic-like, and intrinsic apoptotic triggers, kinases and phosphatases involved in metabolic signal transduction pathways were shown to activate mitochondrial apoptotic pathways after primary murine CECs’ exposure to Aβ peptides. Aβ25–35 activated protein phosphatase 2A (PP2A), a serine/threonine phosphatase with a diverse range of signaling substrates, in primary mouse CECs [190]. It is suggested that PP2A may dephosphorylate and activate ASK1. In this study, ASK1 was shown to activate the MAPK kinase 3 and 6 (MKK3/6) pathways, which activated the p38/MAPK pathway. This led to the activation of p53, which activates Bax and inhibits Bcl-2, and ultimately mitochondria-mediated apoptosis.
Moreover, primary murine CECs’ exposure to Aβ25–35 was shown to downregulate active, phosphorylated forms of Akt and upregulate Bcl-2-associated death promoter (Bad) [191] and Bcl-2-like protein 11 (Bim) [192], which are downstream inhibition targets of Akt. Bad translocated from the cytosol to mitochondria, and inhibited Bcl-XL [191]. The downregulation of Bcl-XL and upregulation of Bim led to release of mitochondrial Smac, which was mediated by the mPTP, eventually allowing activation of caspases and apoptosis to proceed.
Effect of different amyloid peptides
The extent of activation and upregulation of apoptotic pathways involving the mitochondria may vary depending on the Aβ peptide, its rate of oligomerization, and the CEC type. In murine and rat cell lines as well as primary mouse CEC cultures, WT Aβ40, Aβ25–35, and Aβ42 induced mitochondria-mediated apoptosis, upregulation of apoptotic proteins, and mitochondrial DNA damage as described in above subsections. However, by WT Aβ40 amyloid exposure in human CECs showed more delayed effects when compared to vasculotropic mutants associated with CAA and cerebral hemorrhage such as the Dutch (E22Q), Piedmont (L34V), and Iowa (D23N) Aβ40 mutants [117, 146–148, 181].
The speed of amyloid oligomerization matched the speed and extent of apoptosis in hCMEC/D3 cell: Aβ40-E22Q immediately oligomerized and was faster compared to Aβ40-L34V and Aβ40 (WT) [146–148]. Similarly, aggregation kinetics and apoptosis induction were fastest and greatest for Aβ40-D23N (isoD), then Aβ40-D23N, and then Aβ40 (isoD) [117].
This suggests that mutated amyloid peptides more quickly induce mitochondria-mediated apoptosis in CECs than WT peptides, especially Aβ40, which has slower aggregation kinetics compared to Aβ42. Ultimately, all Aβ40 peptides (E22Q, L34V, and WT) induced the activation of caspase-8, which was followed by caspase-9 activation and apoptosis [148]. However, the different kinetics of mitochondrial dysfunction and apoptosis induction clearly reflect the earlier age of disease onset in people possessing these vasculotropic mutations and highlight the relevance of these studies and of mitochondrial dysfunction mechanisms in differentiating between different disease pathologies.
Primary human CEC cultures showed similar results with Aβ40-E22Q but not Aβ42 [181]. While Aβ42 was shown to have the highest tendency to form fibrils, more so than Aβ40-E22Q, only Aβ40-E22Q, and neither Aβ40 (WT) nor Aβ42 (WT), increased apoptosis and triggered cytosolic Bax translocation to and colocalization with the mitochondria and CytC release after 12 and 24 h of exposure [181]. It is possible that this lack of upregulated mitochondrial apoptotic activity may be due to the presence of fibrils, instead of oligomers, in Aβ42 preparations, and to the delayed effect shown by Aβ40 (for which apoptosis and CytC release were only present after 3 day challenge in hCMEC/D3 cells) [117, 146, 148].
CONTRIBUTION OF MITOCHONDRIAL FUNCTION IN NEUROGENESIS
Neurogenesis, or the formation of neurons from neuronal progenitor cells, has been observed in the hippocampus during adulthood and has been proposed as a mechanism for learning and memory formation [193, 194]. Mitochondria have also been shown to play a role in neurogenesis [195, 196]. Therapies that enhance mitochondria bioenergetics such as daily oxaloacetate (OAA) treatment [197, 198], high-intensity exercise [199], and tauroursodeoxycholic acid [200] improve neurogenesis. On the other hand, selective knockout of apoptosis inducing factor during embryonic development induces mitochondrial dysfunction and impairs neurogenesis and cognitive function [201]. Although the mitochondria appear to have an important function in neurogenesis, the role of neurogenesis in AD and cognition, learning, and memory warrants further investigation [202]. For a detailed discussion on the contribution of mitochondrial function in neurogenesis during AD, the reader is directed elsewhere [203–205].
THERAPEUTIC STRATEGIES
Mitochondria-targeted therapies
Recognizing that mitochondria are the main sources of ROS, many mitochondrial-targeted therapies have focused on the development of antioxidant molecules that accumulate in the mitochondrial matrix. One of such molecules is the Szeto-Schiller peptide (SS-31), also known as Bendavia or MPT-131, which accumulates in the mitochondria in a Δψ–independent manner, inhibits mPTP, and serves as a ROS scavenger [206]. The peptide SS-31 was found to prevent age-associated neurovascular uncoupling and mitochondrial dysfunction in microvascular CECs, and this in turn prevented the age-related cognitive decline in WT mice [207]. Although the SS-31 peptide seems to target some of the main features observed in CAA, its potential beneficial effects in cerebral amyloidosis have not been studied.
Another mitochondrial-targeted antioxidant is MitoQ, a Δψ-dependent triphenylphosphonium (TPP+)-ubiquinone derivative that was shown to prevent H2O2-induced apoptosis [208]. This compound was recently approved to undergo clinical trials as a dietary supplement for AD patients where the endpoint is to assess its effects on CBF and carotid endothelial function (). This clinical trial emerged from the observation that mitoQ could reverse vascular defects due to aging, possibly in an NO-dependent manner [209]. It is possible that decreasing ROS levels increases bioavailability of NO by preventing ONOO− formation and restoring vascular function. In addition, in a triple transgenic AD mouse model (3×Tg-AD) of APP with the Swedish mutation, MAPT P301L, and PSEN1 M146V, a five-month treatment with mitoQ prevented cognitive decline, nitrotyrosine formation (a marker of ONOO− damage), synaptic dysfunction, and caspase 3/7 activation [138]. However, other studies have reported adverse effects of MitoQ in the kidney, indicating that MitoQ induced mitochondrial swelling and membrane depolarization due to the increased inner mitochondrial membrane permeability [210]. The effects of MitoQ in reducing ONOO− damage could be especially beneficial in CECs, were it appears that lack of NO bioavailability and excessive ROS production enhance or contribute to the progression of AD.
During the last few years, an FDA-approved drug, the CAI methazolamide, was shown to protect CECs, smooth muscle cells, neurons and glial cells against Aβ-mediated mitochondrial dysfunction and apoptosis [117, 146, 211]. These promising results led our group to also evaluate the effects of the analog CAI acetazolamide on mitochondrial function in cerebral vascular cells as well as neuronal cells, and the mechanisms responsible for the protective effects [111]. Both CAIs were able to prevent H2O2 production, loss of Δψ, activation of caspase 9, and CytC release; thereby inhibiting apoptosis [111]. Due to their FDA-approved use in several medical conditions including glaucoma and high altitude sickness, as well as the potential added benefits in improving CBF and vasoreactivity [212, 213], these compounds could provide a multi-target novel pharmacotherapy for CAA and AD with potential protective effects against cerebrovascular mitochondrial dysfunction.
Multiple studies have focused on the modulation of mitochondrial bioenergetics to prevent cognitive dysfunction. It was reported that the inhibition of Complex I with a small molecule, CP2, is able to prevent the cognitive decline and Aβ accumulation in APP, PS1, and APP/PS1 transgenic mice [214, 215]. Other studies have demonstrated that OAA, can modulate mitochondrial bioenergetics in mice and the human cell line SH-SY5Y [197, 198], reducing neuroinflammation and enhancing the expression of genes involved in neurogenesis [197]. OAA supplementation appears to be a promising therapy since it is well tolerated in humans [216]. These findings further support the mitochondrial as a possible player in neurodegeneration and provide evidence that the regulation of mitochondrial bioenergetics could be developed as a possible therapeutic approach to AD and CAA.
Although less common, some studies have evaluated the use of natural compounds that appear to target the mitochondria directly. One of such compounds is Centella asiatica, an aquatic plant. Water extracts from centella asiatica were able to improve learning and spatial memory in Tg2576 mice (open field test, Morris water maze) without affecting Aβ levels [217]. In contrast to these results, in a different model of AD, PS/APP mice expressing Swedish APP and M146L presenilin 1, a decrease in hippocampal Aβ40 and Aβ42 levels was observed after centella asiatica treatment for 8 months starting at 2 months of age (before the development of amyloid burden) [218]. The effects of centella asiatica might be mediated by a reduction of ROS [218] and preservation of mitochondrial function [219]. However, the actions of centella asiatica on brain microvascular endothelial cells during amyloidosis have not been studied.
Clinical trials and non-pharmacological therapies
Currently, there are no effective cures for AD or CAA. The National Library of Medicine only displays a total of 12 registered clinical trials for CAA that have been or are currently underway (https://clinicaltrials.gov/ct2/results?cond=Cerebral+Amyloid+Angiopathy&term=&cntry=&state=&city=&dist=). However, only three (or 25%) involve a pharmacotherapeutic approach; the vast majority intend to understand the relationships between several risk factors and CAA through imaging studies. Of the three clinical trials involving pharmacotherapy, two phase II clinical trials have been completed which involved an immunotherapy treatment with Ponezumab, which was demonstrated to be inefficient [220, 221], and a glycosaminoglycan mimetic [222] for which the results have not yet been posted.
Interestingly, a recently approved clinical trial will examine the potential effect of oleocanthal rich-extra virgin olive oil (EVOO) on preserving cognitive function and preventing the BBB deficits in mild-cognitive impaired patients. The Mediterranean diet, which is high in EVOO, has been associated to a reduced risk of AD and cognitive impairment [223, 224]. To further explore the potential effects of EVOO against cognitive decline, various in vivo and in vitro studies have been performed. In wild type mice brain endothelial cells, EVOO increased the expression of proteins involved in Aβ40 clearance and this was associated with enhanced clearance of radioactively labeled 125I-Aβ40 after acute injection [225]. Although the 2013 study used WT animals, the same group later demonstrated that in Tg-SwDI mice, treatment with oleocanthal decreased total Aβ accumulation in the hippocampus and brain microvessels; these findings were further confirmed in a human microvascular endothelial cell line [226]. In both studies they demonstrate that the effects of oleocanthal is on Aβ clearance since it does not affect Aβ production. If effective, it would provide a non-pharmacological option to the prevention of cognitive decline and AD. EVOO, known for its antioxidant effects, could also have potential beneficial actions against mitochondrial dysfunction in CAA and AD [227, 228].
Various natural compounds have been associated to beneficial effects in cognitive decline. One of these compounds is Crocus sativus, also known as saffron, a spice that is retrieved from a flower. A phase II clinical trial was held in Iran (Iranian Clinical Trials Registry IRCT138711051556) where 30 mg/kg of saffron a day was able to confer the same beneficial effects of donepezil, but with a marked reduction of the side effects, after 22 days of treatment in mild and moderate cognitive impaired individuals [229]. Crocus supplementation for 12 months improved the Mini-Mental State Examination scores as well as the latency of P300, measurements of cognitive function [230]. Although the clinical results are mild but positive, saffron appears to exert its effects by improving Aβ clearance through upregulation of BBB transporters and prevention of astrocyte activation in 5XFAD mice [231]. However, these studies did not evaluate if these effects improved cognitive function. Lastly, a possible role for physical activity on human cognitive function has been observed. Acute exercise improved pre-frontal but not hippocampal cognitive function [232], while long-term exercise attenuated hippocampal volume decrease and improved memory functions [233]. The beneficial effects resulting from increased physical activity in AD occur at different cellular and metabolic levels, mitochondria being preferential target organelles [234–236]. Moreover, high intensity exercise was shown to increase VEGF-A expression, which induces proliferation and migration of vascular endothelial cells [199].
FUTURE DIRECTIONS
Brain endothelial cells are responsible for regulating BBB function, a process in which mitochondria play an essential role. This is further supported by data demonstrating that brain endothelial cell mitochondria occupy from 8 to 11% of cytoplasmic volume in contrast to non-cerebral endothelium in which mitochondria occupy less than 6% of cytoplasmic volume [101]. Cerebral amyloid angiopathy is characterized by endothelial dysfunction, disruption of the BBB, and vascular damage; all which appear to precede cognitive dysfunction in AD. Therefore, understanding the mechanism by which endothelial dysfunction occurs and the role of the mitochondria in this pathology could provide new therapeutic strategies to prevent AD. However, the role of the mitochondria in endothelial dysfunction during CAA is understudied and further research is needed in multiple directions, among which understanding the mechanisms by which Aβ affects mitochondrial metabolism, respiration, Ca2+ signaling, as well as initiation of mitochondria-mediated apoptotic pathways. Clarifying these pathways will support the development of novel biomarkers and therapeutic approaches for CAA, AD, and vascular mixed dementias.
ACKNOWLEDGMENTS
Financial support was provided by NIH 1R01NS104127.
Authors’ disclosures available online (https://www.j-alz.com/manuscript-disclosures/19-0357r1).
REFERENCES
- [1].Alzheimer’s Association (2019) 2019 Alzheimer’s disease facts and figures. Alzheimers Dement 15, 321–387. [Google Scholar]
- [2].Corriveau RA, Bosetti F, Emr M, Gladman JT, Koenig JI, Moy CS, Pahigiannis K, Waddy SP, Koroshetz W (2016) The science of vascular contributions to cognitive impairment and dementia (VCID): A framework for advancing research priorities in the cerebrovascular biology of cognitive decline. Cell Mol Neurobiol 36, 281–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Attems J, Jellinger KA (2014) The overlap between vascular disease and Alzheimer’s disease–lessons from pathology. BMC Med 12, 206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Jellinger KA, Attems J (2005) Prevalence and pathogenic role of cerebrovascular lesions in Alzheimer disease. J Neurol Sci 229–230, 37–41. [DOI] [PubMed] [Google Scholar]
- [5].Jellinger KA, Attems J (2007) Neuropathological evaluation of mixed dementia. J Neurol Sci 257, 80–87. [DOI] [PubMed] [Google Scholar]
- [6].Jellinger KA, Attems J (2015) Challenges of multimorbidity of the aging brain: A critical update. J Neural Transm (Vienna) 122, 505–521. [DOI] [PubMed] [Google Scholar]
- [7].Schneider JA, Arvanitakis Z, Bang W, Bennett DA (2007) Mixed brain pathologies account for most dementia cases in community-dwelling older persons. Neurology 69, 2197–2204. [DOI] [PubMed] [Google Scholar]
- [8].Snyder HM, Corriveau RA, Craft S, Faber JE, Greenberg SM, Knopman D, Lamb BT, Montine TJ, Nedergaard M, Schaffer CB, Schneider JA, Wellington C, Wilcock DM, Zipfel GJ, Zlokovic B, Bain LJ, Bosetti F, Galis ZS, Koroshetz W, Carrillo MC (2015) Vascular contributions to cognitive impairment and dementia including Alzheimer’s disease. Alzheimers Dement 11, 710–717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Azarpazhooh MR, Avan A, Cipriano LE, Munoz DG, Sposato LA, Hachinski V (2018) Concomitant vascular and neurodegenerative pathologies double the risk of dementia. Alzheimers Dement 14, 148–156. [DOI] [PubMed] [Google Scholar]
- [10].Blennow K, de Leon MJ, Zetterberg H (2006) Alzheimer’s disease. Lancet 368, 387–403. [DOI] [PubMed] [Google Scholar]
- [11].Iadecola C (2010) The overlap between neurodegenerative and vascular factors in the pathogenesis of dementia. Acta Neuropathol 120, 287–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Charidimou A, Boulouis G, Gurol ME, Ayata C, Bacskai BJ, Frosch MP, Viswanathan A, Greenberg SM (2017) Emerging concepts in sporadic cerebral amyloid angiopathy. Brain 140, 1829–1850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Attems J, Yamaguchi H, Saido TC, Thal DR (2010) Capillary CAA and perivascular Abeta-deposition: Two distinct features of Alzheimer’s disease pathology. J Neurol Sci 299, 155–162. [DOI] [PubMed] [Google Scholar]
- [14].Grinberg LT, Thal DR (2010) Vascular pathology in the aged human brain. Acta Neuropathol 119, 277–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Pantoni L (2010) Cerebral small vessel disease: From pathogenesis and clinical characteristics to therapeutic challenges. Lancet Neurol 9, 689–701. [DOI] [PubMed] [Google Scholar]
- [16].Tanskanen M, Makela M, Myllykangas L, Notkola IL, Polvikoski T, Sulkava R, Kalimo H, Paetau A (2012) Prevalence and severity of cerebral amyloid angiopathy: A population-based study on very elderly Finns (Vantaa 85+). Neuropathol Appl Neurobiol 38, 329–336. [DOI] [PubMed] [Google Scholar]
- [17].Biffi A, Greenberg SM (2011) Cerebral amyloid angiopathy: A systematic review. J Clin Neurol 7, 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Ghiso J, Fossati S, Rostagno A (2014) Amyloidosis associated with cerebral amyloid angiopathy: Cell signaling pathways elicited in cerebral endothelial cells. J Alzheimers Dis 42, S167–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Olichney JM, Hansen LA, Hofstetter CR, Lee JH, Katz-man R, Thal LJ (2000) Association between severe cerebral amyloid angiopathy and cerebrovascular lesions in Alzheimer disease is not a spurious one attributable to apolipoprotein E4. Arch Neurol 57, 869–874. [DOI] [PubMed] [Google Scholar]
- [20].Murphy MP, Corriveau RA, Wilcock DM (2016) Vascular contributions to cognitive impairment and dementia (VCID). Biochim Biophys Acta 1862, 857–859. [DOI] [PubMed] [Google Scholar]
- [21].Nation DA, Sweeney MD, Montagne A, Sagare AP, D’Orazio LM, Pachicano M, Sepehrband F, Nelson AR, Buennagel DP, Harrington MG, Benzinger TLS, Fagan AM, Ringman JM, Schneider LS, Morris JC, Chui HC, Law M, Toga AW, Zlokovic BV (2019) Blood-brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nat Med 25, 270–276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Sweeney MD, Montagne A, Sagare AP, Nation DA, Schneider LS, Chui HC, Harrington MG, Pa J, Law M, Wang DJJ, Jacobs RE, Doubal FN, Ramirez J, Black SE, Nedergaard M, Benveniste H, Dichgans M, Iadecola C, Love S, Bath PM, Markus HS, Salman RA, Allan SM, Quinn TJ, Kalaria RN, Werring DJ, Carare RO, Touyz RM, Williams SCR, Moskowitz MA, Katusic ZS, Lutz SE, Lazarov O, Minshall RD, Rehman J, Davis TP, Wellington CL, Gonzalez HM, Yuan C, Lockhart SN, Hughes TM, Chen CLH, Sachdev P, O’Brien JT, Skoog I, Pantoni L, Gustafson DR, Biessels GJ, Wallin A, Smith EE, Mok V, Wong A, Passmore P, Barkof F, Muller M, Breteler MMB, Roman GC, Hamel E, Seshadri S, Gottesman RF, van Buchem MA, Arvanitakis Z, Schneider JA, Drewes LR, Hachinski V, Finch CE, Toga AW, Wardlaw JM, Zlokovic BV (2019) Vascular dysfunction-The disregarded partner of Alzheimer’s disease. Alzheimers Dement 15, 158–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Gorelick PB, Scuteri A, Black SE, Decarli C, Greenberg SM, Iadecola C, Launer LJ, Laurent S, Lopez OL, Nyenhuis D, Petersen RC, Schneider JA, Tzourio C, Arnett DK, Bennett DA, Chui HC, Higashida RT, Lindquist R, Nilsson PM, Roman GC, Sellke FW, Seshadri S, American Heart Association Stroke Council, Council on Epidemiology and Prevention, Council on Cardiovascular Nursing, Council on Cardiovascular Radiology and Intervention, and Council on Cardiovascular Surgery and Anesthesia (2011) Vascular contributions to cognitive impairment and dementia: A statement for healthcare professionals from the american heart association/american stroke association. Stroke 42, 2672–2713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Brown WR, Moody DM, Thore CR, Challa VR (2000) Cerebrovascular pathology in Alzheimer’s disease and leukoaraiosis. Ann N Y Acad Sci 903, 39–45. [DOI] [PubMed] [Google Scholar]
- [25].Brown WR, Moody DM, Thore CR, Challa VR (2000) Apoptosis in leukoaraiosis. AJNR Am J Neuroradiol 21, 79–82. [PMC free article] [PubMed] [Google Scholar]
- [26].Hassan A, Hunt BJ, O’Sullivan M, Bell R, D’Souza R, Jeffery S, Bamford JM, Markus HS (2004) Homocysteine is a risk factor for cerebral small vessel disease, acting via endothelial dysfunction. Brain 127, 212–219. [DOI] [PubMed] [Google Scholar]
- [27].Black S, Gao F, Bilbao J (2009) Understanding white matter disease: Imaging-pathological correlations in vascular cognitive impairment. Stroke 40, S48–52. [DOI] [PubMed] [Google Scholar]
- [28].Gouw AA, Seewann A, van der Flier WM, Barkhof F, Rozemuller AM, Scheltens P, Geurts JJ (2011) Heterogeneity of small vessel disease: A systematic review of MRI and histopathology correlations. J Neurol Neurosurg Psychiatry 82, 126–135. [DOI] [PubMed] [Google Scholar]
- [29].Brown WR, Thore CR (2011) Review: Cerebral microvascular pathology in ageing and neurodegeneration. Neuropathol Appl Neurobiol 37, 56–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].de la Torre JC (2012) Cardiovascular risk factors promote brain hypoperfusion leading to cognitive decline and dementia. Cardiovasc Psychiatry Neurol 2012, 367516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Bink DI, Ritz K, Aronica E, van der Weerd L, Daemen MJ (2013) Mouse models to study the effect of cardiovascular risk factors on brain structure and cognition. J Cereb Blood Flow Metab 33, 1666–1684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Iadecola C (2004) Neurovascular regulation in the normal brain and in Alzheimer’s disease. Nat Rev Neurosci 5, 347–360. [DOI] [PubMed] [Google Scholar]
- [33].Zlokovic BV (2008) The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron 57, 178–201. [DOI] [PubMed] [Google Scholar]
- [34].Schreiber S, Bueche CZ, Garz C, Braun H (2013) Blood brain barrier breakdown as the starting point of cerebral small vessel disease? - New insights from a rat model. Exp Transl Stroke Med 5, 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Scheltens P, Blennow K, Breteler MM, de Strooper B, Frisoni GB, Salloway S, Van der Flier WM (2016) Alzheimer’s disease. Lancet 388, 505–517. [DOI] [PubMed] [Google Scholar]
- [36].Masters CL, Bateman R, Blennow K, Rowe CC, Sperling RA, Cummings JL (2015) Alzheimer’s disease. Nat Rev Dis Primers 1, 15056. [DOI] [PubMed] [Google Scholar]
- [37].Viswanathan A, Greenberg SM (2011) Cerebral amyloid angiopathy in the elderly. Ann Neurol 70, 871–880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Iturria-Medina Y, Sotero RC, Toussaint PJ, Mateos-Perez JM, Evans AC, Alzheimer’s Disease Neuroimaging Initiative (2016) Early role of vascular dysregulation on late-onset Alzheimer’s disease based on multifactorial data-driven analysis. Nat Commun 7, 11934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Xu W, Xu F, Anderson ME, Kotarba AE, Davis J, Robinson JK, Van Nostrand WE (2014) Cerebral microvascular rather than parenchymal amyloid-beta protein pathology promotes early cognitive impairment in transgenic mice. J Alzheimers Dis 38, 621–632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Park L, Koizumi K, El Jamal S, Zhou P, Previti ML, Van Nostrand WE, Carlson G, Iadecola C (2014) Age-dependent neurovascular dysfunction and damage in a mouse model of cerebral amyloid angiopathy. Stroke 45, 1815–1821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Hickman RA, Faustin A, Wisniewski T (2016) Alzheimer disease and its growing epidemic: Risk factors, biomarkers, and the urgent need for therapeutics. Neurol Clin 34, 941–953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Podcasy JL, Epperson CN (2016) Considering sex and gender in Alzheimer disease and other dementias. Dialogues Clin Neurosci 18, 437–446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Barnes DE, Yaffe K (2011) The projected effect of risk factor reduction on Alzheimer’s disease prevalence. Lancet Neurol 10, 819–828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Li J, Wang YJ, Zhang M, Xu ZQ, Gao CY, Fang CQ, Yan JC, Zhou HD, Chongqing Ageing Study Group (2011) Vascular risk factors promote conversion from mild cognitive impairment to Alzheimer disease. Neurology 76, 1485–1491. [DOI] [PubMed] [Google Scholar]
- [45].Shen L, Ji HF (2015) Associations between homocysteine, folic acid, vitamin B12 and Alzheimer’s disease: Insights from meta-analyses. J Alzheimers Dis 46, 777–790. [DOI] [PubMed] [Google Scholar]
- [46].Gustafson D, Rothenberg E, Blennow K, Steen B, Skoog I (2003) An 18-year follow-up of overweight and risk of Alzheimer disease. Arch Intern Med 163, 1524–1528. [DOI] [PubMed] [Google Scholar]
- [47].Arvanitakis Z, Wilson RS, Bienias JL, Evans DA, Bennett DA (2004) Diabetes mellitus and risk of Alzheimer disease and decline in cognitive function. Arch Neurol 61, 661–666. [DOI] [PubMed] [Google Scholar]
- [48].Li W, Risacher SL, McAllister TW, Saykin AJ (2016) Traumatic brain injury and age at onset of cognitive impairment in older adults. J Neurol 263, 1280–1285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Ramos-Cejudo J, Wisniewski T, Marmar C, Zetterberg H, Blennow K, de Leon MJ, Fossati S (2018) Traumatic brain injury and Alzheimer’s disease: The cerebrovascular link. EBioMedicine 28, 21–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Sugarman MA, McKee AC, Stein TD, Tripodis Y, Besser LM, Martin B, Palmisano JN, Steinberg EG, O’Connor MK, Au R, McClean M, Killiany R, Mez J, Weiner MW, Kowall NW, Stern RA, Alosco ML (2019) Failure to detect an association between self-reported traumatic brain injury and Alzheimer’s disease neuropathology and dementia. Alzheimers Dement 15, 686–698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Verghese PB, Castellano JM, Holtzman DM (2011) Apolipoprotein E in Alzheimer’s disease and other neurological disorders. Lancet Neurol 10, 241–252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Guerreiro RJ, Gustafson DR, Hardy J (2012) The genetic architecture of Alzheimer’s disease: Beyond APP, PSENs and APOE. Neurobiol Aging 33, 437–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Bekris LM, Yu CE, Bird TD, Tsuang DW (2010) Genetics of Alzheimer disease. J Geriatr Psychiatry Neurol 23, 213–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Lanoiselee HM, Nicolas G, Wallon D, Rovelet-Lecrux A, Lacour M, Rousseau S, Richard AC, Pasquier F, Rollin-Sillaire A, Martinaud O, Quillard-Muraine M, de la Sayette V, Boutoleau-Bretonniere C, Etcharry-Bouyx F, Chauvire V, Sarazin M, le Ber I, Epelbaum S, Jonveaux T, Rouaud O, Ceccaldi M, Felician O, Godefroy O, Formaglio M, Croisile B, Auriacombe S, Chamard L, Vincent JL, Sauvee M, Marelli-Tosi C, Gabelle A, Ozsancak C, Pariente J, Paquet C, Hannequin D, Campion D, collaborators of the CNR-MAJ project (2017) APP, PSEN1, and PSEN2 mutations in early-onset Alzheimer disease: A genetic screening study of familial and sporadic cases. PLoS Med 14, e1002270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Hatami A, Monjazeb S, Milton S, Glabe CG (2017) Familial Alzheimer’s disease mutations within the amyloid precursor protein alter the aggregation and conformation of the amyloid-beta peptide. J Biol Chem 292, 3172–3185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Haass C, Lemere CA, Capell A, Citron M, Seubert P, Schenk D, Lannfelt L, Selkoe DJ (1995) The Swedish mutation causes early-onset Alzheimer’s disease by beta-secretase cleavage within the secretory pathway. Nat Med 1, 1291–1296. [DOI] [PubMed] [Google Scholar]
- [57].Murrell JR, Hake AM, Quaid KA, Farlow MR, Ghetti B (2000) Early-onset Alzheimer disease caused by a new mutation (V717L) in the amyloid precursor protein gene. Arch Neurol 57, 885–887. [DOI] [PubMed] [Google Scholar]
- [58].Revesz T, Holton JL, Lashley T, Plant G, Frangione B, Rostagno A, Ghiso J (2009) Genetics and molecular pathogenesis of sporadic and hereditary cerebral amyloid angiopathies. Acta Neuropathol 118, 115–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Keable A, Fenna K, Yuen HM, Johnston DA, Smyth NR, Smith C, Al-Shahi Salman R, Samarasekera N, Nicoll JA, Attems J, Kalaria RN, Weller RO, Carare RO (2016) Deposition of amyloid beta in the walls of human leptomeningeal arteries in relation to perivascular drainage pathways in cerebral amyloid angiopathy. Biochim Biophys Acta 1862, 1037–1046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].MacGregor Sharp M, Bulters D, Brandner S, Holton J, Verma A, Werring DJ, Carare RO (2019) The fine anatomy of the perivascular compartment in the human brain: Relevance to dilated perivascular spaces in cerebral amyloid angiopathy. Neuropathol Appl Neurobiol 45, 305–308. [DOI] [PubMed] [Google Scholar]
- [61].Greenberg SM, Charidimou A (2018) Diagnosis of cerebral amyloid angiopathy: Evolution of the Boston criteria. Stroke 49, 491–497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Rostagno A, Holton JL, Lashley T, Revesz T, Ghiso J (2010) Cerebral amyloidosis: Amyloid subunits, mutants and phenotypes. Cell Mol Life Sci 67, 581–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Sweeney MD, Kisler K, Montagne A, Toga AW, Zlokovic BV (2018) The role of brain vasculature in neurodegenerative disorders. Nat Neurosci 21, 1318–1331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Bangen KJ, Nation DA, Delano-Wood L, Weissberger GH, Hansen LA, Galasko DR, Salmon DP, Bondi MW (2015) Aggregate effects of vascular risk factors on cerebrovascular changes in autopsy-confirmed Alzheimer’s disease. Alzheimers Dement 11, 394–403 e391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Kruyer A, Soplop N, Strickland S, Norris EH (2015) Chronic hypertension leads to neurodegeneration in the TgSwDI mouse model of Alzheimer’s disease. Hypertension 66, 175–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Okamoto Y, Yamamoto T, Kalaria RN, Senzaki H, Maki T, Hase Y, Kitamura A, Washida K, Yamada M, Ito H, Tomimoto H, Takahashi R, Ihara M (2012) Cerebral hypoperfusion accelerates cerebral amyloid angiopathy and promotes cortical microinfarcts. Acta Neuropathol 123, 381–394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].de la Torre JC (2002) Vascular basis of Alzheimer’s pathogenesis. Ann N Y Acad Sci 977, 196–215. [DOI] [PubMed] [Google Scholar]
- [68].Rius-Perez S, Tormos AM, Perez S, Talens-Visconti R (2018) Vascular pathology: Cause or effect in Alzheimer disease? Neurologia 33, 112–120. [DOI] [PubMed] [Google Scholar]
- [69].Kitaguchi H, Tomimoto H, Ihara M, Shibata M, Uemura K, Kalaria RN, Kihara T, Asada-Utsugi M, Kinoshita A, Takahashi R (2009) Chronic cerebral hypoperfusion accelerates amyloid beta deposition in APPSwInd transgenic mice. Brain Res 1294, 202–210. [DOI] [PubMed] [Google Scholar]
- [70].Cai Z, Liu Z, Xiao M, Wang C, Tian F (2017) Chronic cerebral hypoperfusion promotes amyloid-beta pathogenesis via activating beta/gamma-secretases. Neurochem Res 42, 3446–3455. [DOI] [PubMed] [Google Scholar]
- [71].Salvadores N, Searcy JL, Holland PR, Horsburgh K (2017) Chronic cerebral hypoperfusion alters amyloid-beta peptide pools leading to cerebral amyloid angiopathy, microinfarcts and haemorrhages in Tg-SwDI mice. Clin Sci (Lond) 131, 2109–2123. [DOI] [PubMed] [Google Scholar]
- [72].Bannai T, Mano T, Chen X, Ohtomo G, Ohtomo R, Tsuchida T, Koshi-Mano K, Hashimoto T, Okazawa H, Iwatsubo T, Tsuji S, Toda T, Iwata A (2019) Chronic cerebral hypoperfusion shifts the equilibrium of amyloid beta oligomers to aggregation-prone species with higher molecular weight. Sci Rep 9, 2827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Zhang X, Zhou K, Wang R, Cui J, Lipton SA, Liao FF, Xu H, Zhang YW (2007) Hypoxia-inducible factor 1alpha (HIF-1alpha)-mediated hypoxia increases BACE1 expression and beta-amyloid generation. J Biol Chem 282, 10873–10880. [DOI] [PubMed] [Google Scholar]
- [74].Bulbarelli A, Lonati E, Brambilla A, Orlando A, Cazzaniga E, Piazza F, Ferrarese C, Masserini M, Sancini G (2012) Abeta42 production in brain capillary endothelial cells after oxygen and glucose deprivation. Mol Cell Neurosci 49, 415–422. [DOI] [PubMed] [Google Scholar]
- [75].Correia SC, Moreira PI (2010) Hypoxia-inducible factor 1: A new hope to counteract neurodegeneration? J Neurochem 112, 1–12. [DOI] [PubMed] [Google Scholar]
- [76].Huang Y, Lei L, Liu D, Jovin I, Russell R, Johnson RS, Di Lorenzo A, Giordano FJ (2012) Normal glucose uptake in the brain and heart requires an endothelial cell-specific HIF-1alpha-dependent function. Proc Natl Acad Sci U S A 109, 17478–17483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Iyalomhe O, Swierczek S, Enwerem N, Chen Y, Adedeji MO, Allard J, Ntekim O, Johnson S, Hughes K, Kurian P, Obisesan TO (2017) The role of hypoxia-inducible factor 1 in mild cognitive impairment. Cell Mol Neurobiol 37, 969–977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Hawkins BT, Davis TP (2005) The blood-brain barrier/neurovascular unit in health and disease. Pharmacol Rev 57, 173–185. [DOI] [PubMed] [Google Scholar]
- [79].Sweeney MD, Sagare AP, Zlokovic BV (2018) Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat Rev Neurol 14, 133–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Zlokovic BV (2011) Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat Rev Neurosci 12, 723–738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Kalaria RN, Hedera P (1995) Differential degeneration of the cerebral microvasculature in Alzheimer’s disease. Neuroreport 6, 477–480. [DOI] [PubMed] [Google Scholar]
- [82].Claudio L (1996) Ultrastructural features of the blood-brain barrier in biopsy tissue from Alzheimer’s disease patients. Acta Neuropathol 91, 6–14. [DOI] [PubMed] [Google Scholar]
- [83].van de Haar HJ, Burgmans S, Jansen JF, van Osch MJ, van Buchem MA, Muller M, Hofman PA, Verhey FR, Backes WH (2016) Blood-brain barrier leakage in patients with early Alzheimer disease. Radiology 281, 527–535. [DOI] [PubMed] [Google Scholar]
- [84].Montagne A, Zhao Z, Zlokovic BV (2017) Alzheimer’s disease: A matter of blood-brain barrier dysfunction? J Exp Med 214, 3151–3169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Luissint AC, Artus C, Glacial F, Ganeshamoorthy K, Couraud PO (2012) Tight junctions at the blood brain barrier: Physiological architecture and disease-associated dysregulation. Fluids Barriers CNS 9, 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Hartz AM, Bauer B, Soldner EL, Wolf A, Boy S, Backhaus R, Mihaljevic I, Bogdahn U, Klunemann HH, Schuierer G, Schlachetzki F (2012) Amyloid-beta contributes to blood-brain barrier leakage in transgenic human amyloid precursor protein mice and in humans with cerebral amyloid angiopathy. Stroke 43, 514–523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Yamazaki Y, Shinohara M, Shinohara M, Yamazaki A, Murray ME, Liesinger AM, Heckman MG, Lesser ER, Parisi JE, Petersen RC, Dickson DW, Kanekiyo T, Bu G (2019) Selective loss of cortical endothelial tight junction proteins during Alzheimer’s disease progression. Brain 142, 1077–1092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Balietti M, Giorgetti B, Casoli T, Solazzi M, Tamagnini F, Burattini C, Aicardi G, Fattoretti P (2013) Early selective vulnerability of synapses and synaptic mitochondria in the hippocampal CA1 region of the Tg2576 mouse model of Alzheimer’s disease. J Alzheimers Dis 34, 887–896. [DOI] [PubMed] [Google Scholar]
- [89].Du H, Guo L, Yan S, Sosunov AA, McKhann GM, Yan SS (2010) Early deficits in synaptic mitochondria in an Alzheimer’s disease mouse model. Proc Natl Acad Sci U S A 107, 18670–18675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Manczak M, Anekonda TS, Henson E, Park BS, Quinn J, Reddy PH (2006) Mitochondria are a direct site of A beta accumulation in Alzheimer’s disease neurons: Implications for free radical generation and oxidative damage in disease progression. Hum Mol Genet 15, 1437–1449. [DOI] [PubMed] [Google Scholar]
- [91].Santos RX, Correia SC, Wang X, Perry G, Smith MA, Moreira PI, Zhu X (2010) A synergistic dysfunction of mitochondrial fission/fusion dynamics and mitophagy in Alzheimer’s disease. J Alzheimers Dis 20(Suppl 2), S401–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Swerdlow RH, Khan SM (2004) A “mitochondrial cascade hypothesis” for sporadic Alzheimer’s disease. Med Hypotheses 63, 8–20. [DOI] [PubMed] [Google Scholar]
- [93].Moreira PI, Carvalho C, Zhu X, Smith MA, Perry G (2010) Mitochondrial dysfunction is a trigger of Alzheimer’s disease pathophysiology. Biochim Biophys Acta 1802, 2–10. [DOI] [PubMed] [Google Scholar]
- [94].Varghese M, Zhao W, Wang J, Cheng A, Qian X, Chaudhry A, Ho L, Pasinetti GM (2011) Mitochondrial bioenergetics is defective in presymptomatic Tg2576 AD mice. Transl Neurosci 2. doi: 10.2478/s13380-011-0011-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Yao J, Irwin RW, Zhao L, Nilsen J, Hamilton RT, Brinton RD (2009) Mitochondrial bioenergetic deficit precedes Alzheimer’s pathology in female mouse model of Alzheimer’s disease. Proc Natl Acad Sci U S A 106, 14670–14675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Burguillos MA, Deierborg T, Kavanagh E, Persson A, Hajji N, Garcia-Quintanilla A, Cano J, Brundin P, Englund E, Venero JL, Joseph B (2011) Caspase signalling controls microglia activation and neurotoxicity. Nature 472, 319–324. [DOI] [PubMed] [Google Scholar]
- [97].D’Amelio M, Cavallucci V, Middei S, Marchetti C, Pacioni S, Ferri A, Diamantini A, De Zio D, Carrara P, Battistini L, Moreno S, Bacci A, Ammassari-Teule M, Marie H, Cecconi F (2011) Caspase-3 triggers early synaptic dysfunction in a mouse model of Alzheimer’s disease. Nat Neurosci 14, 69–76. [DOI] [PubMed] [Google Scholar]
- [98].de Calignon A, Fox LM, Pitstick R, Carlson GA, Bacskai BJ, Spires-Jones TL, Hyman BT (2010) Caspase activation precedes and leads to tangles. Nature 464, 1201–1204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Eckert A, Marques CA, Keil U, Schussel K, Muller WE (2003) Increased apoptotic cell death in sporadic and genetic Alzheimer’s disease. Ann N Y Acad Sci 1010, 604–609. [DOI] [PubMed] [Google Scholar]
- [100].Nikolaev A, McLaughlin T, O’Leary DD, Tessier-Lavigne M (2009) APP binds DR6 to trigger axon pruning and neuron death via distinct caspases. Nature 457, 981–989. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- [101].Oldendorf WH, Cornford ME, Brown WJ (1977) The large apparent work capability of the blood-brain barrier: A study of the mitochondrial content of capillary endothelial cells in brain and other tissues of the rat. Ann Neurol 1, 409–417. [DOI] [PubMed] [Google Scholar]
- [102].Nguyen HM, Mejia EM, Chang W, Wang Y, Watson E, On N, Miller DW, Hatch GM (2016) Reduction in cardiolipin decreases mitochondrial spare respiratory capacity and increases glucose transport into and across human brain cerebral microvascular endothelial cells. J Neurochem 139, 68–80. [DOI] [PubMed] [Google Scholar]
- [103].Belanger M, Allaman I, Magistretti PJ (2011) Brain energy metabolism: Focus on astrocyte-neuron metabolic cooperation. Cell Metab 14, 724–738. [DOI] [PubMed] [Google Scholar]
- [104].Zheng PP, Romme E, van der Spek PJ, Dirven CM, Willemsen R, Kros JM (2010) Glut1/SLC2A1 is crucial for the development of the blood-brain barrier in vivo. Ann Neurol 68, 835–844. [DOI] [PubMed] [Google Scholar]
- [105].Yin KJ, Lee JM, Chen SD, Xu J, Hsu CY (2002) Amyloid-beta induces Smac release via AP-1/Bim activation in cerebral endothelial cells. J Neurosci 22, 9764–9770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Casley CS, Land JM, Sharpe MA, Clark JB, Duchen MR, Canevari L (2002) Beta-amyloid fragment 25–35 causes mitochondrial dysfunction in primary cortical neurons. Neurobiol Dis 10, 258–267. [DOI] [PubMed] [Google Scholar]
- [107].Casley CS, Canevari L, Land JM, Clark JB, Sharpe MA (2002) Beta-amyloid inhibits integrated mitochondrial respiration and key enzyme activities. J Neurochem 80, 91–100. [DOI] [PubMed] [Google Scholar]
- [108].Bobba A, Amadoro G, Valenti D, Corsetti V, Lassandro R, Atlante A (2013) Mitochondrial respiratory chain Complexes I and IV are impaired by beta-amyloid via direct interaction and through Complex I-dependent ROS production, respectively. Mitochondrion 13, 298–311. [DOI] [PubMed] [Google Scholar]
- [109].Hernandez-Zimbron LF, Luna-Munoz J, Mena R, Vazquez-Ramirez R, Kubli-Garfias C, Cribbs DH, Manoutcharian K, Gevorkian G (2012) Amyloid-beta peptide binds to cytochrome C oxidase subunit 1. PLoS One 7, e42344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Schmidt C, Lepsverdize E, Chi SL, Das AM, Pizzo SV, Dityatev A, Schachner M (2008) Amyloid precursor protein and amyloid beta-peptide bind to ATP synthase and regulate its activity at the surface of neural cells. Mol Psychiatry 13, 953–969. [DOI] [PubMed] [Google Scholar]
- [111].Solesio ME, Peixoto PM, Debure L, Madamba SM, de Leon MJ, Wisniewski T, Pavlov EV, Fossati S (2018) Carbonic anhydrase inhibition selectively prevents amyloid beta neurovascular mitochondrial toxicity. Aging Cell, e12787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Newington JT, Pitts A, Chien A, Arseneault R, Schubert D, Cumming RC (2011) Amyloid beta resistance in nerve cell lines is mediated by the Warburg effect. PLoS One 6, e19191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Murphy MP (2009) How mitochondria produce reactive oxygen species. Biochem J 417, 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Turrens JF (2003) Mitochondrial formation of reactive oxygen species. J Physiol 552, 335–344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [115].Badhwar A, Brown R, Stanimirovic DB, Haqqani AS, Hamel E (2017) Proteomic differences in brain vessels of Alzheimer’s disease mice: Normalization by PPARgamma agonist pioglitazone. J Cereb Blood Flow Metab 37, 1120–1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].Han BH, Zhou ML, Johnson AW, Singh I, Liao F, Vellimana AK, Nelson JW, Milner E, Cirrito JR, Basak J, Yoo M, Dietrich HH, Holtzman DM, Zipfel GJ (2015) Contribution of reactive oxygen species to cerebral amyloid angiopathy, vasomotor dysfunction, and microhemorrhage in aged Tg2576 mice. Proc Natl Acad Sci U S A 112, E881–890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [117].Fossati S, Todd K, Sotolongo K, Ghiso J, Rostagno A (2013) Differential contribution of isoaspartate post-translational modifications to the fibrillization and toxic properties of amyloid beta and the Asn23 Iowa mutation. Biochem J 456, 347–360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].Zhao L, Hou L, Sun H, Yan X, Sun X, Li J, Bian Y, Chu Y, Liu Q (2011) Apigenin isolated from the medicinal plant Elsholtzia rugulosa prevents β-amyloid 25–35-induces toxicity in rat cerebral microvascular endothelial cells. Molecules 16, 4005–4019. [Google Scholar]
- [119].Liu T, Liu WH, Zhao JS, Meng FZ, Wang H (2017) Lutein protects against beta-amyloid peptide-induced oxidative stress in cerebrovascular endothelial cells through modulation of Nrf-2 and NF-kappab. Cell Biol Toxicol 33, 57–67. [DOI] [PubMed] [Google Scholar]
- [120].Fonseca AC, Moreira PI, Oliveira CR, Cardoso SM, Pinton P, Pereira CF (2015) Amyloid-beta disrupts calcium and redox homeostasis in brain endothelial cells. Mol Neurobiol 51, 610–622. [DOI] [PubMed] [Google Scholar]
- [121].Bates TE, Loesch A, Burnstock G, Clark JB (1995) Immunocytochemical evidence for a mitochondrially located nitric oxide synthase in brain and liver. Biochem Biophys Res Commun 213, 896–900. [DOI] [PubMed] [Google Scholar]
- [122].Ghafourifar P, Cadenas E (2005) Mitochondrial nitric oxide synthase. Trends Pharmacol Sci 26, 190–195. [DOI] [PubMed] [Google Scholar]
- [123].Brookes PS (2004) Mitochondrial nitric oxide synthase. Mitochondrion 3, 187–204. [DOI] [PubMed] [Google Scholar]
- [124].Shiva S, Castro L, Brookes PS (2017) Biology and pathobiology In Nitric Oxide, Ignarro LJ, Freeman BA, eds. Academic Press, pp. 137–156. [Google Scholar]
- [125].Chisari M, Merlo S, Sortino MA, Salomone S (2010) Long-term incubation with beta-amyloid peptides impairs endothelium-dependent vasodilatation in isolated rat basilar artery. Pharmacol Res 61, 157–161. [DOI] [PubMed] [Google Scholar]
- [126].Suo Z, Humphrey J, Kundtz A, Sethi F, Placzek A, Crawford F, Mullan M (1998) Soluble Alzheimers beta-amyloid constricts the cerebral vasculature in vivo. Neurosci Lett 257, 77–80. [DOI] [PubMed] [Google Scholar]
- [127].Jeynes B, Provias J (2009) Significant negative correlations between capillary expressed eNOS and Alzheimer lesion burden. Neurosci Lett 463, 244–248. [DOI] [PubMed] [Google Scholar]
- [128].Austin SA, Santhanam AV, Hinton DJ, Choi DS, Katusic ZS (2013) Endothelial nitric oxide deficiency promotes Alzheimer’s disease pathology. J Neurochem 127, 691–700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].Tan XL, Xue YQ, Ma T, Wang X, Li JJ, Lan L, Malik KU, McDonald MP, Dopico AM, Liao FF (2015) Partial eNOS deficiency causes spontaneous thrombotic cerebral infarction, amyloid angiopathy and cognitive impairment. Mol Neurodegener 10, 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Keil U, Bonert A, Marques CA, Strosznajder JB, Muller-Spahn F, Muller WE, Eckert A (2004) Elevated nitric oxide production mediates beta-amyloid-induced mitochondria failure. Pol J Pharmacol 56, 631–634. [PubMed] [Google Scholar]
- [131].Radi R (2013) Peroxynitrite, a stealthy biological oxidant. J Biol Chem 288, 26464–26472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Radi R, Cassina A, Hodara R, Quijano C, Castro L (2002) Peroxynitrite reactions and formation in mitochondria. Free Radic Biol Med 33, 1451–1464. [DOI] [PubMed] [Google Scholar]
- [133].Xia Y, Tsai AL, Berka V, Zweier JL (1998) Superoxide generation from endothelial nitric-oxide synthase. A Ca2+/calmodulin-dependent and tetrahydrobiopterin regulatory process. J Biol Chem 273, 25804–25808. [DOI] [PubMed] [Google Scholar]
- [134].Massaad CA, Amin SK, Hu L, Mei Y, Klann E, Pautler RG (2010) Mitochondrial superoxide contributes to blood flow and axonal transport deficits in the Tg2576 mouse model of Alzheimer’s disease. PLoS One 5, e10561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].Good PF, Werner P, Hsu A, Olanow CW, Perl DP (1996) Evidence of neuronal oxidative damage in Alzheimer’s disease. Am J Pathol 149, 21–28. [PMC free article] [PubMed] [Google Scholar]
- [136].Smith MA, Richey Harris PL, Sayre LM, Beckman JS, Perry G (1997) Widespread peroxynitrite-mediated damage in Alzheimer’s disease. J Neurosci 17, 2653–2657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Park L, Anrather J, Forster C, Kazama K, Carlson GA, Iadecola C (2004) Abeta-induced vascular oxidative stress and attenuation of functional hyperemia in mouse somatosensory cortex. J Cereb Blood Flow Metab 24, 334–342. [DOI] [PubMed] [Google Scholar]
- [138].McManus MJ, Murphy MP, Franklin JL (2011) The mitochondria-targeted antioxidant MitoQ prevents loss of spatial memory retention and early neuropathology in a transgenic mouse model of Alzheimer’s disease. J Neurosci 31, 15703–15715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Merlini M, Shi Y, Keller S, Savarese G, Akhmedov A, Derungs R, Spescha RD, Kulic L, Nitsch RM, Luscher TF, Camici GG (2017) Reduced nitric oxide bioavailability mediates cerebroarterial dysfunction independent of cerebral amyloid angiopathy in a mouse model of Alzheimer’s disease. Am J Physiol Heart Circ Physiol 312, H232–H238. [DOI] [PubMed] [Google Scholar]
- [140].Duran-Prado M, Frontinan J, Santiago-Mora R, Peinado JR, Parrado-Fernandez C, Gomez-Almagro MV, Moreno M, Lopez-Dominguez JA, Villalba JM, Alcain FJ (2014) Coenzyme Q10 protects human endothelial cells from beta-amyloid uptake and oxidative stress-induced injury. PLoS One 9, e109223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [141].Katakam PV, Wappler EA, Katz PS, Rutkai I, Institoris A, Domoki F, Gaspar T, Grovenburg SM, Snipes JA, Busija DW (2013) Depolarization of mitochondria in endothelial cells promotes cerebral artery vasodilation by activation of nitric oxide synthase. Arterioscler Thromb Vasc Biol 33, 752–759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Bolanos JP, Peuchen S, Heales SJ, Land JM, Clark JB (1994) Nitric oxide-mediated inhibition of the mitochondrial respiratory chain in cultured astrocytes. J Neurochem 63, 910–916. [DOI] [PubMed] [Google Scholar]
- [143].Cleeter MW, Cooper JM, Darley-Usmar VM, Moncada S, Schapira AH (1994) Reversible inhibition of cytochrome c oxidase, the terminal enzyme of the mitochondrial respiratory chain, by nitric oxide. Implications for neurodegenerative diseases. FEBS Lett 345, 50–54. [DOI] [PubMed] [Google Scholar]
- [144].Guivernau B, Bonet J, Valls-Comamala V, Bosch-Morato M, Godoy JA, Inestrosa NC, Peralvarez-Marin A, Fernandez-Busquets X, Andreu D, Oliva B, Munoz FJ (2016) Amyloid-beta peptide nitrotyrosination stabilizes oligomers and enhances NMDAR-mediated toxicity. J Neurosci 36, 11693–11703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [145].Verma M, Vats A, Taneja V (2015) Toxic species in amyloid disorders: Oligomers or mature fibrils. Ann Indian Acad Neurol 18, 138–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [146].Fossati S, Cam J, Meyerson J, Mezhericher E, Romero IA, Couraud PO, Weksler BB, Ghiso J, Rostagno A (2010) Differential activation of mitochondrial apoptotic pathways by vasculotropic amyloid-beta variants in cells composing the cerebral vessel walls. FASEB J 24, 229–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [147].Fossati S, Ghiso J, Rostagno A (2012) Insights into caspase-mediated apoptotic pathways induced by amyloid-beta in cerebral microvascular endothelial cells. Neurodegener Dis 10, 324–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [148].Fossati S, Ghiso J, Rostagno A (2012) TRAIL death receptors DR4 and DR5 mediate cerebral microvascular endothelial cell apoptosis induced by oligomeric Alzheimer’s Abeta. Cell Death Dis 3, e321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [149].Kummer MP, Hermes M, Delekarte A, Hammerschmidt T, Kumar S, Terwel D, Walter J, Pape HC, Konig S, Roeber S, Jessen F, Klockgether T, Korte M, Heneka MT (2011) Nitration of tyrosine 10 critically enhances amyloid beta aggregation and plaque formation. Neuron 71, 833–844. [DOI] [PubMed] [Google Scholar]
- [150].Paris D, Parker TA, Town T, Suo Z, Fang C, Humphrey J, Crawford F, Mullan M (1998) Role of peroxynitrite in the vasoactive and cytotoxic effects of Alzheimer’s beta-amyloid1–40 peptide. Exp Neurol 152, 116–122. [DOI] [PubMed] [Google Scholar]
- [151].Pacher P, Beckman JS, Liaudet L (2007) Nitric oxide and peroxynitrite in health and disease. Physiol Rev 87, 315–424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [152].Denton RM (2009) Regulation of mitochondrial dehydrogenases by calcium ions. Biochim Biophys Acta 1787, 1309–1316. [DOI] [PubMed] [Google Scholar]
- [153].Momoi T (2004) Caspases involved in ER stress-mediated cell death. J Chem Neuroanat 28, 101–105. [DOI] [PubMed] [Google Scholar]
- [154].Magi S, Castaldo P, Macri ML, Maiolino M, Matteucci A, Bastioli G, Gratteri S, Amoroso S, Lariccia V (2016) Intracellular calcium dysregulation: Implications for Alzheimer’s disease. Biomed Res Int 2016, 6701324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [155].Ferreiro E, Oliveira CR, Pereira CM (2008) The release of calcium from the endoplasmic reticulum induced by amyloid-beta and prion peptides activates the mitochondrial apoptotic pathway. Neurobiol Dis 30, 331–342. [DOI] [PubMed] [Google Scholar]
- [156].Fonseca AC, Ferreiro E, Oliveira CR, Cardoso SM, Pereira CF (2013) Activation of the endoplasmic reticulum stress response by the amyloid-beta 1–40 peptide in brain endothelial cells. Biochim Biophys Acta 1832, 2191–2203. [DOI] [PubMed] [Google Scholar]
- [157].Cioffi DL, Stevens T (2006) Regulation of endothelial cell barrier function by store-operated calcium entry. Microcirculation 13, 709–723. [DOI] [PubMed] [Google Scholar]
- [158].Park L, Wang G, Moore J, Girouard H, Zhou P, Anrather J, Iadecola C (2014) The key role of transient receptor potential melastatin-2 channels in amyloid-beta-induced neurovascular dysfunction. Nat Commun 5, 5318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [159].Xu J, Chen S, Ku G, Ahmed SH, Xu J, Chen H, Hsu CY (2001) Amyloid beta peptide-induced cerebral endothelial cell death involves mitochondrial dysfunction and caspase activation. J Cereb Blood Flow Metab 21, 702–710. [DOI] [PubMed] [Google Scholar]
- [160].Abramov AY, Fraley C, Diao CT, Winkfein R, Colicos MA, Duchen MR, French RJ, Pavlov E (2007) Targeted polyphosphatase expression alters mitochondrial metabolism and inhibits calcium-dependent cell death. Proc Natl Acad Sci U S A 104, 18091–18096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [161].Redza-Dutordoir M, Averill-Bates DA (2016) Activation of apoptosis signalling pathways by reactive oxygen species. Biochim Biophys Acta 1863, 2977–2992. [DOI] [PubMed] [Google Scholar]
- [162].Roos WP, Thomas AD, Kaina B (2016) DNA damage and the balance between survival and death in cancer biology. Nat Rev Cancer 16, 20–33. [DOI] [PubMed] [Google Scholar]
- [163].Pihan P, Carreras-Sureda A, Hetz C (2017) BCL-2 family: Integrating stress responses at the ER to control cell demise. Cell Death Differ 24, 1478–1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [164].Chang F, Lee JT, Navolanic PM, Steelman LS, Shelton JG, Blalock WL, Franklin RA, McCubrey JA (2003) Involvement of PI3K/Akt pathway in cell cycle progression, apoptosis, and neoplastic transformation: A target for cancer chemotherapy. Leukemia 17, 590–603. [DOI] [PubMed] [Google Scholar]
- [165].Hatai T, Matsuzawa A, Inoshita S, Mochida Y, Kuroda T, Sakamaki K, Kuida K, Yonehara S, Ichijo H, Takeda K (2000) Execution of apoptosis signal-regulating kinase 1 (ASK1)-induced apoptosis by the mitochondria-dependent caspase activation. J Biol Chem 275, 26576–26581. [DOI] [PubMed] [Google Scholar]
- [166].Chen Z, Seimiya H, Naito M, Mashima T, Kizaki A, Dan S, Imaizumi M, Ichijo H, Miyazono K, Tsuruo T (1999) ASK1 mediates apoptotic cell death induced by genotoxic stress. Oncogene 18, 173–180. [DOI] [PubMed] [Google Scholar]
- [167].Dhanasekaran DN, Reddy EP (2008) JNK signaling in apoptosis. Oncogene 27, 6245–6251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [168].Elmore S (2007) Apoptosis: A review of programmed cell death. Toxicol Pathol 35, 495–516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [169].Kagan VE, Tyurin VA, Jiang J, Tyurina YY, Ritov VB, Amoscato AA, Osipov AN, Belikova NA, Kapralov AA, Kini V, Vlasova II, Zhao Q, Zou M, Di P, Svistunenko DA, Kurnikov IV, Borisenko GG (2005) Cytochrome c acts as a cardiolipin oxygenase required for release of proapoptotic factors. Nat Chem Biol 1, 223–232. [DOI] [PubMed] [Google Scholar]
- [170].Korytowski W, Basova LV, Pilat A, Kernstock RM, Girotti AW (2011) Permeabilization of the mitochondrial outer membrane by Bax/truncated Bid (tBid) proteins as sensitized by cardiolipin hydroperoxide translocation: Mechanistic implications for the intrinsic pathway of oxidative apoptosis. J Biol Chem 286, 26334–26343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [171].Degterev A, Boyce M, Yuan J (2003) A decade of caspases. Oncogene 22, 8543–8567. [DOI] [PubMed] [Google Scholar]
- [172].Kandimalla KK, Scott OG, Fulzele S, Davidson MW, Poduslo JF (2009) Mechanism of neuronal versus endothelial cell uptake of Alzheimer’s disease amyloid beta protein. PLoS One 4, e4627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [173].Fonseca AC, Oliveira CR, Pereira CF, Cardoso SM (2014) Loss of proteostasis induced by amyloid beta peptide in brain endothelial cells. Biochim Biophys Acta 1843, 1150–1161. [DOI] [PubMed] [Google Scholar]
- [174].Solito R, Corti F, Chen CH, Mochly-Rosen D, Giachetti A, Ziche M, Donnini S (2013) Mitochondrial aldehyde dehydrogenase-2 activation prevents beta-amyloid-induced endothelial cell dysfunction and restores angiogenesis. J Cell Sci 126, 1952–1961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [175].Singh Angom R, Wang Y, Wang E, Pal K, Bhattacharya S, Watzlawik JO, Rosenberry TL, Das P, Mukhopadhyay D (2019) VEGF receptor-1 modulates amyloid beta 1–42 oligomer-induced senescence in brain endothelial cells. FASEB J 33, 4626–4637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [176].Ferreira ST, Lourenco MV, Oliveira MM, De Felice FG (2015) Soluble amyloid-beta oligomers as synaptotoxins leading to cognitive impairment in Alzheimer’s disease. Front Cell Neurosci 9, 191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [177].Fulda S, Debatin KM (2006) Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy. Oncogene 25, 4798–4811. [DOI] [PubMed] [Google Scholar]
- [178].Bhardwaj A, Aggarwal BB (2003) Receptor-mediated choreography of life and death. J Clin Immunol 23, 317–332. [DOI] [PubMed] [Google Scholar]
- [179].Guicciardi ME, Gores GJ (2009) Life and death by death receptors. FASEB J 23, 1625–1637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [180].Haase G, Pettmann B, Raoul C, Henderson CE (2008) Signaling by death receptors in the nervous system. Curr Opin Neurobiol 18, 284–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [181].Viana RJ, Nunes AF, Castro RE, Ramalho RM, Meyerson J, Fossati S, Ghiso J, Rostagno A, Rodrigues CM (2009) Tauroursodeoxycholic acid prevents E22Q Alzheimer’s Abeta toxicity in human cerebral endothelial cells. Cell Mol Life Sci 66, 1094–1104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [182].Chen W, Chan Y, Wan W, Li Y, Zhang C (2018) Abeta1–42 induces cell damage via RAGE-dependent endoplasmic reticulum stress in bEnd.3 cells. Exp Cell Res 362, 83–89. [DOI] [PubMed] [Google Scholar]
- [183].Park L, Wang G, Zhou P, Zhou J, Pitstick R, Previti ML, Younkin L, Younkin SG, Van Nostrand WE, Cho S, Anrather J, Carlson GA, Iadecola C (2011) Scavenger receptor CD36 is essential for the cerebrovascular oxidative stress and neurovascular dysfunction induced by amyloid-beta. Proc Natl Acad Sci U S A 108, 5063–5068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [184].Sessa L, Gatti E, Zeni F, Antonelli A, Catucci A, Koch M, Pompilio G, Fritz G, Raucci A, Bianchi ME (2014) The receptor for advanced glycation end-products (RAGE) is only present in mammals, and belongs to a family of cell adhesion molecules (CAMs). PLoS One 9, e86903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [185].Yan SD, Chen X, Fu J, Chen M, Zhu H, Roher A, Slattery T, Zhao L, Nagashima M, Morser J, Migheli A, Nawroth P, Stern D, Schmidt AM (1996) RAGE and amyloid-beta peptide neurotoxicity in Alzheimer’s disease. Nature 382, 685–691. [DOI] [PubMed] [Google Scholar]
- [186].Candela P, Gosselet F, Saint-Pol J, Sevin E, Boucau MC, Boulanger E, Cecchelli R, Fenart L (2010) Apical-to-basolateral transport of amyloid-beta peptides through blood-brain barrier cells is mediated by the receptor for advanced glycation end-products and is restricted by P-glycoprotein. J Alzheimers Dis 22, 849–859. [DOI] [PubMed] [Google Scholar]
- [187].Smith LM, Kostylev MA, Lee S, Strittmatter SM (2019) Systematic and standardized comparison of reported amyloid-beta receptors for sufficiency, affinity, and Alzheimer’s disease relevance. J Biol Chem 294, 6042–6053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [188].Mitchell RW, On NH, Del Bigio MR, Miller DW, Hatch GM (2011) Fatty acid transport protein expression in human brain and potential role in fatty acid transport across human brain microvessel endothelial cells. J Neurochem 117, 735–746. [DOI] [PubMed] [Google Scholar]
- [189].Silverstein RL, Febbraio M (2009) CD36, a scavenger receptor involved in immunity, metabolism, angiogenesis, and behavior. Sci Signal 2, re3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [190].Hsu MJ, Hsu CY, Chen BC, Chen MC, Ou G, Lin CH (2007) Apoptosis signal-regulating kinase 1 in amyloid beta peptide-induced cerebral endothelial cell apoptosis. J Neurosci 27, 5719–5729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [191].Yin KJ, Lee JM, Chen H, Xu J, Hsu CY (2005) Abeta25–35 alters Akt activity, resulting in Bad translocation and mitochondrial dysfunction in cerebrovascular endothelial cells. J Cereb Blood Flow Metab 25, 1445–1455. [DOI] [PubMed] [Google Scholar]
- [192].Yin KJ, Hsu CY, Hu XY, Chen H, Chen SW, Xu J, Lee JM (2006) Protein phosphatase 2A regulates bim expression via the Akt/FKHRL1 signaling pathway in amyloid-beta peptide-induced cerebrovascular endothelial cell death. J Neurosci 26, 2290–2299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [193].Aimone JB, Li Y, Lee SW, Clemenson GD, Deng W, Gage FH (2014) Regulation and function of adult neurogenesis: From genes to cognition. Physiol Rev 94, 991–1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [194].Goncalves JT, Schafer ST, Gage FH (2016) Adult neurogenesis in the hippocampus: From stem cells to behavior. Cell 167, 897–914. [DOI] [PubMed] [Google Scholar]
- [195].Voloboueva LA, Lee SW, Emery JF, Palmer TD, Giffard RG (2010) Mitochondrial protection attenuates inflammation-induced impairment of neurogenesis in vitro and in vivo. J Neurosci 30, 12242–12251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [196].Arrazola MS, Andraini T, Szelechowski M, Mouledous L, Arnaune-Pelloquin L, Davezac N, Belenguer P, Rampon C, Miquel MC (2018) Mitochondria in developmental and adult neurogenesis. Neurotox Res. doi: 10.1007/s12640-018-9942-y [DOI] [PubMed] [Google Scholar]
- [197].Wilkins HM, Harris JL, Carl SM, E L, Lu J, Eva Selfridge J, Roy N, Hutfles L, Koppel S, Morris J, Burns JM, Michaelis ML, Michaelis EK, Brooks WM, Swerdlow RH (2014) Oxaloacetate activates brain mitochondrial biogenesis, enhances the insulin pathway, reduces inflammation and stimulates neurogenesis. Hum Mol Genet 23, 6528–6541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [198].Wilkins HM, Koppel S, Carl SM, Ramanujan S, Weidling I, Michaelis ML, Michaelis EK, Swerdlow RH (2016) Oxaloacetate enhances neuronal cell bioenergetic fluxes and infrastructure. J Neurochem 137, 76–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [199].E L, Burns JM, Swerdlow RH (2014) Effect of high-intensity exercise on aged mouse brain mitochondria, neurogenesis, and inflammation. Neurobiol Aging 35, 2574–2583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [200].Soares R, Ribeiro FF, Xapelli S, Genebra T, Ribeiro MF, Sebastiao AM, Rodrigues CMP, Sola S (2018) Tauroursodeoxycholic acid enhances mitochondrial biogenesis, neural stem cell pool, and early neurogenesis in adult rats. Mol Neurobiol 55, 3725–3738. [DOI] [PubMed] [Google Scholar]
- [201].Khacho M, Clark A, Svoboda DS, MacLaurin JG, Lagace DC, Park DS, Slack RS (2017) Mitochondrial dysfunction underlies cognitive defects as a result of neural stem cell depletion and impaired neurogenesis. Hum Mol Genet 26, 3327–3341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [202].Martinez-Canabal A (2014) Reconsidering hippocampal neurogenesis in Alzheimer’s disease. Front Neurosci 8, 147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [203].Bartolome F, de la Cueva M, Pascual C, Antequera D, Fernandez T, Gil C, Martinez A, Carro E (2018) Amyloid beta-induced impairments on mitochondrial dynamics, hippocampal neurogenesis, and memory are restored by phosphodiesterase 7 inhibition. Alzheimers Res Ther 10,24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [204].Dard RF, Dahan L, Rampon C (2018) Targeting hippocampal adult neurogenesis using transcription factors to reduce Alzheimer’s disease-associated memory impairments. Hippocampus. doi: 10.1002/hipo.23052 [DOI] [PubMed] [Google Scholar]
- [205].Lazarov O, Hollands C (2016) Hippocampal neurogenesis: Learning to remember. Prog Neurobiol 138–140, 1–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [206].Zhao K, Zhao GM, Wu D, Soong Y, Birk AV, Schiller PW, Szeto HH (2004) Cell-permeable peptide antioxidants targeted to inner mitochondrial membrane inhibit mitochondrial swelling, oxidative cell death, and reperfusion injury. J Biol Chem 279, 34682–34690. [DOI] [PubMed] [Google Scholar]
- [207].Tarantini S, Valcarcel-Ares NM, Yabluchanskiy A, Fulop GA, Hertelendy P, Gautam T, Farkas E, Perz A, Rabinovitch PS, Sonntag WE, Csiszar A, Ungvari Z (2018) Treatment with the mitochondrial-targeted antioxidant peptide SS-31 rescues neurovascular coupling responses and cerebrovascular endothelial function and improves cognition in aged mice. Aging Cell 17, e12731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [208].Kelso GF, Porteous CM, Coulter CV, Hughes G, Porteous WK, Ledgerwood EC, Smith RA, Murphy MP (2001) Selective targeting of a redox-active ubiquinone to mitochondria within cells: Antioxidant and antiapoptotic properties. J Biol Chem 276, 4588–4596. [DOI] [PubMed] [Google Scholar]
- [209].Park SY, Kwon OS, Andtbacka RHI, Hyngstrom JR, Reese V, Murphy MP, Richardson RS (2018) Age-related endothelial dysfunction in human skeletal muscle feed arteries: The role of free radicals derived from mitochondria in the vasculature. Acta Physiol (Oxf) 222, e12893. [DOI] [PubMed] [Google Scholar]
- [210].Gottwald EM, Duss M, Bugarski M, Haenni D, Schuh CD, Landau EM, Hall AM (2018) The targeted anti-oxidant MitoQ causes mitochondrial swelling and depolarization in kidney tissue. Physiol Rep 6, e13667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [211].Fossati S, Giannoni P, Solesio ME, Cocklin SL, Cabrera E, Ghiso J, Rostagno A (2016) The carbonic anhydrase inhibitor methazolamide prevents amyloid beta-induced mitochondrial dysfunction and caspase activation protecting neuronal and glial cells in vitro and in the mouse brain. Neurobiol Dis 86, 29–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [212].Okudaira Y, Bandoh K, Arai H, Sato K (1995) Evaluation of the acetazolamide test. Vasoreactivity and cerebral blood volume. Stroke 26, 1234–1239. [DOI] [PubMed] [Google Scholar]
- [213].Ulrich S, Nussbaumer-Ochsner Y, Vasic I, Hasler E, Latshang TD, Kohler M, Muehlemann T, Wolf M, Bloch KE (2014) Cerebral oxygenation in patients with OSA: Effects of hypoxia at altitude and impact of acetazolamide. Chest 146, 299–308. [DOI] [PubMed] [Google Scholar]
- [214].Zhang L, Zhang S, Maezawa I, Trushin S, Minhas P, Pinto M, Jin LW, Prasain K, Nguyen TD, Yamazaki Y, Kanekiyo T, Bu G, Gateno B, Chang KO, Nath KA, Nemutlu E, Dzeja P, Pang YP, Hua DH, Trushina E (2015) Modulation of mitochondrial complex I activity averts cognitive decline in multiple animal models of familial Alzheimer’s Disease. EBioMedicine 2, 294–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [215].Swerdlow RH (2015) Mitochondrial manipulation and the quest for Alzheimer’s treatments. EBioMedicine 2, 276–277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [216].Swerdlow RH, Bothwell R, Hutfles L, Burns JM, Reed GA (2016) Tolerability and pharmacokinetics of oxaloacetate 100 mg capsules in Alzheimer’s subjects. BBA Clin 5, 120–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [217].Soumyanath A, Zhong YP, Henson E, Wadsworth T, Bishop J, Gold BG, Quinn JF (2012) Centella asiatica extract improves behavioral deficits in a mouse model of Alzheimer’s disease: Investigation of a possible mechanism of action. Int J Alzheimers Dis 2012, 381974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [218].Dhanasekaran M, Holcomb LA, Hitt AR, Tharakan B, Porter JW, Young KA, Manyam BV (2009) Centella asiatica extract selectively decreases amyloid beta levels in hippocampus of Alzheimer’s disease animal model. Phytother Res 23, 14–19. [DOI] [PubMed] [Google Scholar]
- [219].Gray NE, Zweig JA, Caruso M, Zhu JY, Wright KM, Quinn JF, Soumyanath A (2018) Centella asiatica attenuates hippocampal mitochondrial dysfunction and improves memory and executive function in beta-amyloid overexpressing mice. Mol Cell Neurosci 93, 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [220].Landen JW, Cohen S, Billing CB Jr., Cronenberger C, Styren S, Burstein AH, Sattler C, Lee JH, Jack CR Jr., Kantarci K, Schwartz PF, Duggan WT, Zhao Q, Sprenger K, Bednar MM, Binneman B (2017) Multiple-dose ponezumab for mild-to-moderate Alzheimer’s disease: Safety and efficacy. Alzheimers Dement (N Y) 3, 339–347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [221].Landen JW, Andreasen N, Cronenberger CL, Schwartz PF, Borjesson-Hanson A, Ostlund H, Sattler CA, Binneman B, Bednar MM (2017) Ponezumab in mild-to-moderate Alzheimer’s disease: Randomized phase II PET-PIB study. Alzheimers Dement (N Y) 3, 393–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [222].Gervais F, Chalifour R, Garceau D, Kong X, Laurin J, McLaughlin R, Morissette C, Paquette J (2001) Glycosaminoglycan mimetics: A therapeutic approach to cerebral amyloid angiopathy. Amyloid 8(Suppl 1), 28–35. [PubMed] [Google Scholar]
- [223].Lourida I, Soni M, Thompson-Coon J, Purandare N, Lang IA, Ukoumunne OC, Llewellyn DJ (2013) Mediterranean diet, cognitive function, and dementia: A systematic review. Epidemiology 24, 479–489. [DOI] [PubMed] [Google Scholar]
- [224].Petersson SD, Philippou E (2016) Mediterranean diet, cognitive function, and dementia: A systematic review of the evidence. Adv Nutr 7, 889–904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [225].Abuznait AH, Qosa H, Busnena BA, El Sayed KA, Kaddoumi A (2013) Olive-oil-derived oleocanthal enhances beta-amyloid clearance as a potential neuroprotective mechanism against Alzheimer’s disease: In vitro and in vivo studies. ACS Chem Neurosci 4, 973–982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [226].Qosa H, Batarseh YS, Mohyeldin MM, El Sayed KA, Keller JN, Kaddoumi A (2015) Oleocanthal enhances amyloid-beta clearance from the brains of TgSwDI mice and in vitro across a human blood-brain barrier model. ACS Chem Neurosci 6, 1849–1859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [227].Oliveras-Lopez MJ, Molina JJ, Mir MV, Rey EF, Martin F, de la Serrana HL (2013) Extra virgin olive oil (EVOO) consumption and antioxidant status in healthy institutionalized elderly humans. Arch Gerontol Geriatr 57, 234–242. [DOI] [PubMed] [Google Scholar]
- [228].Lauretti E, Iuliano L, Pratico D (2017) Extra-virgin olive oil ameliorates cognition and neuropathology of the 3×Tg mice: Role of autophagy. Ann Clin Transl Neurol 4, 564–574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [229].Akhondzadeh S, Shafiee Sabet M, Harirchian MH, Togha M, Cheraghmakani H, Razeghi S, Hejazi SS, Yousefi MH, Alimardani R, Jamshidi A, Rezazadeh SA, Yousefi A, Zare F, Moradi A, Vossoughi A (2010) A 22-week, multicenter, randomized, double-blind controlled trial of Crocus sativus in the treatment of mild-to-moderate Alzheimer’s disease. Psychopharmacology (Berl) 207, 637–643. [DOI] [PubMed] [Google Scholar]
- [230].Tsolaki M, Karathanasi E, Lazarou I, Dovas K, Verykouki E, Karacostas A, Georgiadis K, Tsolaki A, Adam K, Kompatsiaris I, Sinakos Z (2016) Efficacy and safety of Crocus sativus L. in patients with mild cognitive impairment: One year single-blind randomized, with parallel groups, clinical trial. J Alzheimers Dis 54, 129–133. [DOI] [PubMed] [Google Scholar]
- [231].Batarseh YS, Bharate SS, Kumar V, Kumar A, Vishwakarma RA, Bharate SB, Kaddoumi A (2017) Crocus sativus extract tightens the blood-brain barrier, reduces amyloid beta load and related toxicity in 5XFAD mice. ACS Chem Neurosci 8, 1756–1766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [232].Basso JC, Shang A, Elman M, Karmouta R, Suzuki WA (2015) Acute exercise improves prefrontal cortex but not hippocampal function in healthy adults. J Int Neuropsychol Soc 21, 791–801. [DOI] [PubMed] [Google Scholar]
- [233].Erickson KI, Voss MW, Prakash RS, Basak C, Szabo A, Chaddock L, Kim JS, Heo S, Alves H, White SM, Wojcicki TR, Mailey E, Vieira VJ, Martin SA, Pence BD, Woods JA, McAuley E, Kramer AF (2011) Exercise training increases size of hippocampus and improves memory. Proc Natl Acad Sci U S A 108, 3017–3022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [234].Bernardo TC, Marques-Aleixo I, Beleza J, Oliveira PJ, Ascensao A, Magalhaes J (2016) Physical exercise and brain mitochondrial fitness: The possible role against Alzheimer’s disease. Brain Pathol 26, 648–663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [235].Ansari A, Rahman MS, Saha SK, Saikot FK, Deep A, Kim KH (2017) Function of the SIRT3 mitochondrial deacetylase in cellular physiology, cancer, and neurodegenerative disease. Aging Cell 16, 4–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [236].Bo H, Kang W, Jiang N, Wang X, Zhang Y, Ji LL (2014) Exercise-induced neuroprotection of hippocampus in APP/PS1 transgenic mice via upregulation of mitochondrial 8-oxoguanine DNA glycosylase. Oxid Med Cell Longev 2014, 834502. [DOI] [PMC free article] [PubMed] [Google Scholar]