Skip to main content

Chronic cerebral hypoperfusion: a critical feature in unravelling the etiology of vascular cognitive impairment

Abstract

Vascular cognitive impairment (VCI) describes a wide spectrum of cognitive deficits related to cerebrovascular diseases. Although the loss of blood flow to cortical regions critically involved in cognitive processes must feature as the main driver of VCI, the underlying mechanisms and interactions with related disease processes remain to be fully elucidated. Recent clinical studies of cerebral blood flow measurements have supported the role of chronic cerebral hypoperfusion (CCH) as a major driver of the vascular pathology and clinical manifestations of VCI. Here we review the pathophysiological mechanisms as well as neuropathological changes of CCH. Potential interventional strategies for VCI are also reviewed. A deeper understanding of how CCH can lead to accumulation of VCI-associated pathology could potentially pave the way for early detection and development of disease-modifying therapies, thus allowing preventive interventions instead of symptomatic treatments.

Introduction

A key missing piece in dementia research is the elucidation of the neurovascular basis of cognitive impairment [230, 258, 308]. The term vascular cognitive impairment (VCI) is used to describe a wide spectrum of conditions characterized by cerebrovascular disease ranging from subjective cognitive decline to vascular dementia (VaD) (Fig. 1). While VaD remains the second most common type of dementia worldwide after Alzheimer’s disease (AD), its prevalence may be underestimated—especially in populations with significant concomitant small vessel disease burden, such as those in Asia [42, 48, 49]. Vascular factors may also exacerbate the pathology of AD [221], giving rise to the argument that vascular pathology could even be the most common contributor to dementia in elderly populations [222]. Moreover, VaD is associated with a high mortality rate and rapid stepwise disease progression (Fig. 1) [4, 5, 116]. Therefore, the identification of interventions to potentially benefit VCI patients and reduce its socioeconomic burden is of critical importance.

Fig. 1
figure 1

Stepwise progression to VaD. The road from risk factors to disease manifestation in VCI is a complicated one, as the multiple demographics, lifestyle and comorbid disease risk and mitigating factors interact through the progression from asymptomatic vascular lesions, cognitive impairment, and finally to VaD. Furthermore, these complex interactions give rise to several distinct cerebrovascular diseases underlying different forms of vascular brain injuries, leading to the clinical heterogeneity of VaD. However, regardless of the specific nature of vascular injury (occlusive, thrombotic, etc.), a state of chronic cerebral hypoperfusion can be considered to be the common etiological link. *See Table 1 for details and summary of supporting research

The pathophysiology and clinical characteristics of VCI have been extensively reviewed [99, 281, 283, 304]. Briefly, VCI is characterized by brain lesions that occur due to vascular pathology which leads to diverse cognitive impairments. These lesions result in ischemic, hemorrhagic, and hypoperfusive states that can manifest as various clinical symptoms. Such vascular pathophysiological states in turn lead to a range of downstream effects and structural changes on the brain, including infarcts, lacunes, microbleeds, white matter injury and parenchymal lesions [108, 139, 140, 147, 228, 247]. VCI thus exists as a heterogeneous group of diseases which can be divided into various subtypes, such as multi-infarct dementia, post-stroke dementia and subcortical ischemic vascular dementia, each having unique features that may manifest clinically as dementia over time. While these disease subtypes provide an avenue to categorize the differing disease etiologies, they have a common set of risk factors, including demographic factors, lifestyle factors or presence of co-morbid conditions (summarized in Table 1). Each of these risk factors is known to independently contribute to the progression of cerebrovascular disease and is therefore associated with cognitive dysfunction and impact the progression to dementia. While there are several treatment options to manage VCI and their underlying risk factors as highlighted in Table 2, the root causes of the problem remain unclear. Hence, there remains several knowledge gaps in the understanding of VCI pathophysiology. Although several mechanisms have been reported to have roles in VCI progression, including neuroinflammation, oxidative stress-induced brain damage, neurodegeneration and brain atrophy [254], we still lack a thorough understanding of this complex disease, and even the aforementioned mechanisms have not been fully elucidated. Furthermore, the clinical signs and symptoms of VCI vary between patients given the heterogeneity of the severity and site of injury. Hence, a consensus on the underlying causes of VCI needs to be reached.

Table 1 Risk factors for vascular dementia (VaD)
Table 2 Current pharmacotherapeutic options for vascular dementia (VaD)

Chronic cerebral hypoperfusion (CCH) refers to chronically inadequate brain perfusion [56]. Given that CCH is intimately associated with various risk factors, pathophysiological processes and pathological lesions known to be involved in VCI (Fig. 1), we propose that CCH is the central underlying cause for the progression of VCI. We emphasize CCH as the underlying cause as it ties together some of the known mechanisms of VCI such as chronic inflammation, oxidative stress, neurodegeneration and brain atrophy. Age-related vascular changes lead to a state of global CCH and induce pathophysiological changes such as blood–brain barrier dysfunction, resulting in increased vulnerability to disease even in the absence of risk factors [278]. Recently, CCH was identified as the common feature observed in multiple subtypes of VCI [74, 283]. Furthermore, it was reported that global cerebral blood flow was significantly lower in VaD than in age-matched controls [227, 231] but significantly higher than in AD [227]. The reductions in cerebral blood flow is one of the earliest features observed from early VCI to VaD [130, 145, 224], and is consistently observed in different brain regions, such as a reported 31% decrease in cerebral blood flow in the frontal cortex and a 39% decrease in the parietal cortex [234]. Compromised cerebral blood flow in the deep white matter of the brain is also associated with hemodynamic ischemic injury, and therefore leads to a higher volume of white matter lesions (WMLs) [18, 286]. Moreover, cerebral hypoperfusion was shown to be a good predictor for WMLs in VCI patients [176, 198]. Given the above evidence, we postulate that CCH is a common driver of VCI pathologies such as WMLs, lacunes, infarcts, and subsequent cognitive impairment.

CCH is strongly associated with stepwise cognitive decline in VCI, where much of what is known about its clinical manifestation along the spectrum from normal to end-stage VCI comes from multiple longitudinal studies involving recruited subjects [128, 135, 200, 224, 268, 275, 286]. Neuropathological evidence, neuropsychological assessments and imaging are important adjuncts in many of these studies to ensure accurate study recruitment. In this review, we explore the concept that CCH is the main mediator of VCI pathology and cognitive impairment. We highlight the links between mechanisms and the development of structural neuropathological changes during VCI and provide a landscape of how each of these changes lead to the development of cognitive impairment in patients. Understanding how CCH drives VCI progression from pre-clinical to severe dementia is essential for both researchers and clinicians in diagnosing and developing novel therapeutics for early intervention.

Pathophysiology of chronic cerebral hypoperfusion (CCH)

In VCI, isolated instances of vascular injury may accumulate into widespread damage that overcome intrinsic repair mechanisms in areas critical for cognitive functions. There are several possible mechanisms that govern the transition from a physiological to a pathological state in the brain during cerebral vascular disease. In order to understand how this transition occurs, animal studies employing CCH are commonly used to model the underlying pathology of VCI. Mouse CCH models are generally generated by manipulating the common carotid artery, for instance, via bilateral common carotid artery stenosis [242] or asymmetric common carotid artery surgery [114], both of which could lead to reduced cerebral blood flow, white matter rarefaction, glial activation, as well as subsequent cognitive impairment. A cascade of molecular and cellular events has been shown to be involved in the pathogenesis of CCH including energy imbalance, oxidative stress, endoplasmic reticulum stress, mitochondrial dysfunction and inflammation (Fig. 2).

Fig. 2
figure 2

Pathological drivers of CCH-associated VCI. Several CCH-induced pathological drivers have been long associated with the pathogenesis of VaD including energy imbalance, inflammation, endoplasmic reticulum (ER) stress, oxidative stress and mitochondrial dysfunction. Decreased ATP production impairs ATPase pumps, results in neuronal depolarization, and leads to a deregulation in the glutamate homeostasis at the synaptic cleft and excitotoxicity in the brain. Low cerebral blood flow triggers the brain to utilize anaerobic respiration to produce ATP and this results in the accumulation of lactate within the neurons, leading to acidosis. Neuronal death following CCH is primarily attributed to the increase in the pro-inflammatory cytokine release during the chronic inflammatory response. Danger associated molecular patterns (DAMPs) released by the brain cells can also trigger glial activation and leukocyte infiltration, both of which can also produce pro-inflammatory cytokines. At the cellular level, increase in the reactive oxidative species from various sources, including the mitochondria, induces the oxidative stress state. While the increase in reactive oxygen species can contribute to the redox dynamics and hemodynamics imbalance, it can also induce chronic ER stress. Persistent ER stress leads to an accumulation of misfolded proteins and can have fatal effects on neuronal survival and integrity via the terminal unfolded protein response (UPR) pathway, as well as contributing to Ca2+ homeostatic imbalance. Mitochondrial deterioration causes a further decrease in the ATP production, leading to proteosomal dysfunction, as well as contributing to the frequency of mutagenesis events at the mitochondrial DNA. In a chronic state of CCH, these drivers are pathological and ultimately pave the way for downstream disease mechanisms

Pathogenic mechanisms

Energy imbalance

During VCI, blood vessels in the brain including arterioles, veins and capillaries are partially occluded or hypoperfused [34, 145, 234]. Disruption to glucose and oxygen supply compromises the production of adenosine triphosphate (ATP) [118, 119]. The resultant state of energy imbalance impairs the function of ATP-dependent sodium–potassium pumps [82] which are critical for maintaining the resting membrane potential of neurons. As such, neurons spontaneously depolarize and release the excitatory neurotransmitter glutamate into the synaptic cleft. The excess accumulation of glutamate in the synaptic cleft is exacerbated by other defective ion pumps that fail to recycle the glutamate, leading to persistent depolarization and overstimulation of neighboring neurons [14, 38]. This excessive activation of glutamate receptors (i.e. NMDA and AMPA receptors) due to energy imbalance that results in neuronal dysfunction and death is called excitotoxicity, which has been reported to occur in chronic diseases such as VCI [265]. In order to compensate for the lack of glucose, the brain will begin to undergo anaerobic glycolysis, which produces lactate. Accumulation of lactate in the brain in turn leads to acidosis and acidotoxicity [143, 301].

Oxidative stress

Oxidative stress is defined as an environment where pro-oxidant species dominates over anti-oxidant species [95]. It is one of the central drivers of pathology in many diseases and it is implicated in the cognitive decline in VCI [26, 53, 166, 172]. Correspondingly, a reduction of circulatory antioxidant enzyme levels (e.g., superoxide dismutase, catalase) and antioxidant capacity (e.g., glutathione, ergothioneine) have been observed in VCI patients [80, 241, 297, 298]. In the brain, CCH causes a disruption in calcium (Ca2+) homeostasis which leads to acute and chronic production of reactive oxygen species [82, 166] from various sources, including electron transport chain, nicotinamide adenine dinucleotide phosphate oxidases (Nox) and nitric oxide synthase. In animal models of CCH, reduction of endothelial nitric oxide synthase expression [189] disrupts the vascular tone and exacerbates cerebral blood flow hypoperfusion [83]. CCH also increases Nox-1 expression in neurons, inducing apoptosis and contributing to cognitive impairment [53]. Oxidative stress also increases levels of circulating nitric oxide synthase inhibitor, reducing nitric oxide bioavailability, leading to vasodilation impairment as evident in cognitive impairment [67]. As the stiffness and pulsatility of the vessels increase, higher sheer stress is generated which disrupts normal continuous blood flow [19]. These vascular changes have been reported to be associated with reduced blood supply to white matter regions, thus precipitating the formation of white matter lesions and lacunes [266, 293].

Endoplasmic reticulum stress

Endoplasmic reticulum stress is emerging as a pathological mechanism in the etiology of VCI [195]. The endoplasmic reticulum is involved in the synthesis and post-translational modifications of molecules that are important in maintaining Ca2+ homeostasis [300]. Being the site of translation, protein folding and transport, disruptions to endoplasmic reticulum’s physiological function in the form of endoplasmic reticulum-calcium depletion, hypoxic conditions and oxidative stress, are known to result in misfolding and accumulation of unfolded integral proteins. Such stressors to endoplasmic reticulum activate an adaptive stress response pathway known as the unfolded protein response (UPR) [229]. This pathway involves three independent endoplasmic reticulum membrane-associated sensors which are protein kinase R-like endoplasmic reticulum kinase (PERK), inositol-requiring protein 1 (IRE1) and activating transcription factor 6 (ATF6) [300].

Under prolonged endoplasmic reticulum stress, cellular proteostasis becomes unsustainable, resulting in accumulation of misfolded proteins and activation of terminal UPR [120]. Attenuation of endoplasmic reticulum stress-induced apoptosis has been found to confer protection against ischemia and reperfusion injury [296]. More specifically, studies using neuronal models of vascular dementia have shown the contribution of zinc-induced neurotoxicity to its pathogenesis, upregulating endoplasmic reticulum stress-related genes like CCAAT-enhancer-binding protein homologous protein (CHOP) and growth-arrest- and DNA-damage-inducible gene 34 (GADD34) [263]. The same group also found that the endoplasmic reticulum stress pathway is involved in zinc-induced neurotoxicity thus implying its possible roles as both a cause and consequence in driving VaD [144].

Mitochondrial dysfunction

Given the high energy demands of the brain, mitochondria, as the ‘powerhouse of the cell’, play a central role in producing energy in the form of ATP. Mitochondria are also vital in regulating brain cell survival and death by controlling the movement of calcium ions between the cells and the extracellular surroundings. Reactive oxygen species produced by the mitochondrial energy-redox axis can signal for apoptosis when the cells are damaged [104, 159, 196, 218].

Mitochondrial dysfunction results in decreased energy production, thus altering cellular redox dynamics in the brain. Under these conditions, mitochondria begin producing an excess of \({\text{O}}_{2}^{ \cdot - }\) and H2O2 molecules in response to increased oxidation of proteins, phospholipids and DNA that pushes the redox equilibrium towards a pro-oxidative state [192]. There is also a global reduction in mitochondrial protein complexes over time [104, 159, 161, 310]. Therefore, it is not surprising that mitochondrial dysfunction has been observed in VaD [162]. In particular, mitochondrial damage such as increased mitochondrial bioenergetic deficits in the hippocampus plays important roles in the spatial learning and memory decline in both human patients and in CCH rodent models [16, 72, 162, 172]. Defects in mitochondrial metabolism lead to altered patterns in the mitochondrial respiratory rate, altered membrane potential, decreased pyruvate hydrogenase levels, increased oxidative stress as manifested by increased hydrogen peroxidase levels. However, mitochondrial dysfunction may not occur independently of other pathological processes but is commonly observed to overlap with other mechanisms such as oxidative stress and proteasome dysfunction [70].

Mitochondrial DNA contains genes that encode the cell’s mitochondrial energy production machinery, and defects or mutations in the mitochondrial DNA have been associated with age-related dementia and neuropathology [29, 31, 68, 102, 287]. The m.3316G > A mutation has been identified in early-onset VaD patients who did not manifest typical vascular symptoms [155]. Rather, the mutation causes a reduction in the activity of the respiratory chain complex I, and hence is associated with the well-established link that cerebrovascular damage increases when mitochondrial energy chain complexes are compromised [125].

Neuroinflammation

Inflammation involves a complex range of responses that are known to play a role in disease conditions. While inflammation is important in tissue repair and recovery, under disease conditions, chronic activation of inflammatory responses results in a destructive phenotype that is observed during disease development and progression [62]. Both acute and chronic inflammation have been implicated in cellular injury associated with a hypoxic state of VCI [136, 174, 217]. Under CCH, reduced blood supply disturbs cellular integrity, activates glial cells and recruits peripheral immune cells to the brain [23, 141, 309], causing death of neighboring cells and secondary tissue damage. Various molecular mechanisms such as activation of inflammatory pathways and inflammasome activation have been shown to play a role in inflammation during CCH.

Systemic inflammation serves as the initial signal of a stressed cell involving the release of damage associated molecular patterns (DAMPs), which are recognized by the pattern recognition receptors, namely Toll-like receptors (TLR) and NOD-like receptors (NLR) on neighboring cells, initiating an inflammatory response [82, 260]. Further studies have established the association of NLR family pyrin domain containing 3 (NLRP3) and Absent in melanoma-2 (AIM2), both of which are involved in inflammasome activation, with VaD and CCH [81, 177, 206, 207]. Regulatory pathways such as nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) are subsequently activated to upregulate a wide range of inflammatory proteins [105, 146] including interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF) [24, 317]. These inflammatory cytokines can cause cell death, oligodendrocyte damage and demyelination [264, 305]. Attenuation of IL-1β production was shown to ameliorate hypoperfusion-induced brain injury in mice [206]. Microglia and astrocytes also release adhesion molecules and chemokines, which activate and facilitate leukocyte infiltration [15, 20, 115]. In a mouse model of cerebral ischemia, genetic deletion of the chemokine CCL2 has been shown to reduce brain injury via modulation of inflammation. Other proinflammatory proteins such as c-reactive protein are also upregulated to facilitate cerebral inflammation in VCI patients [77, 215, 232].

The complement system has also been implicated in stroke [168]. Complement proteins promote inflammation via glial activation and induce neuronal injury through the C5 activating membrane attack complex (MAC). Formation and deposition of C5b-9/MAC complexes damages the myelin sheath [175, 233], and abrogation of C5 protein reduces glial activation and white matter ischemia under CCH [167]. The central effector protein in the system is the C3 convertase enzyme complex [28, 197, 240]. It has been demonstrated that under CCH, microglial cells aggravate white matter injury via the C3-C3aR pathway in rat brains [311]. Together, the entire inflammatory process facilitates astrogliosis and scar formation, oligodendrocyte and endothelial cell dysfunction and blood–brain barrier disruption [248, 282, 313], leading to neurodegeneration, neurovascular dissociation and eventually structural damage to the brain. Therefore, inflammation serves as a critical mechanism that drives subsequent pathological changes in CCH. In summary, the pathological mechanisms of CCH covered in the above section lay a foundation to comprehend the complex mechanistic underpinnings of the disease. The pathological mechanisms of CCH include the involvement of multiple molecules and signaling pathways, especially those related to inflammation and oxidative stress.

Neuropathological features of CCH

In this section, we will examine how the pathogenic mechanisms described above contribute to the neuropathological features that have been described in VCI.

Glial activation

The term neurovascular unit describes the structural and functional interactions between neurons, glial cells, pericytes, extracellular matrix components and endothelial cells in the brain. The neurovascular unit maintains homeostasis within the brain microenvironment ensuring optimal conditions for function of neurons and other cells. During CCH, the entire neurovascular unit is affected by the combined effects of the pathological mechanisms described above that can cause reduced integrity of the neurovascular unit [238]. This results in a homeostatic imbalance in the brain.

Glial cells, especially microglia, drive inflammatory responses by releasing proinflammatory molecules. Increased number of glial cells are commonly seen in VCI patients especially at the white matter regions [245, 269]. Mechanistic studies using animal models have shown that upon CCH, activated microglial cells participated in both systemic and complement-activated inflammation; whereas attenuation of microglial activity reduces proinflammatory cytokine levels, increases myelin density and eventually improves cognitive performance [138, 311].

Astrocytes are also involved in the process of inflammation during CCH. Astrogliosis has direct influence on blood–brain barrier integrity and induces damage when constitutively activated astrocytes form glial scars or swelling at the end feet processes [88, 211]. With CCH, a study reported decreased astrocyte polarity and structural support to the endothelial cells eventually contributing to blood–brain barrier damage [127].

In the white matter, oligodendrocytes are the predominant glial cell type, and produce the myelin sheath around myelinated axons. As CCH damages oligodendrocytes and white matter, repair mechanisms are often impaired due to inflammation and loss of growth factors released by neurons, microglia and astrocytes. The myelin-independent axonal support from oligodendrocyte is also affected, causing significant axonal loss [93, 181, 313]. Upon ischemia, oligodendrocytes also release inhibitory proteins Nogo-A and MMP-9, preventing neuronal remodeling, and initiating a deleterious cascade within white matter to cause blood–brain barrier damage [93, 181].

Together, dysfunction in each of the components within the neurovascular unit can result in the disruption of brain homeostasis, which can eventually lead to neuronal loss and white matter infarctions at the grey matter and the deep white matter territory [35, 252].

Activation of cell death

Programmed cell death is a critical role in animal development and tissue homeostasis. Abnormal regulation of programmed cell death is associated with various human diseases including neurodegeneration. Different forms of cell death such as apoptosis, pyroptosis and autophagy have been observed in cerebral ischemia and reperfusion injury [76, 84, 141]. Of these, there has been ample evidence in the literature implicating apoptosis in VCI. In postmortem studies of VCI patients, apoptotic vascular cells were identified in the basal ganglia and subcortical white matter regions [103]. Apoptotic neuronal cells were also observed at cortical layers 3 and 5; and extensive ischemic lesions and axonal damage were observed in severe dementia [103]. Furthermore, protein expression and proteomics studies have revealed a decreasing anti-apoptotic proteins expression pattern in the cortex of VCI patients compared to controls [64]. Within regions of leukoaraiosis, significant increases in apoptotic oligodendrocytes were observed compared to adjacent white matter [33]. Mirroring the evidence seen in human patients, animal models of CCH present similar results, with increased markers of apoptosis observed. Specific changes observed in these animal studies include increased visualization of apoptotic bodies, increased expression of apoptotic proteins such as caspase 3, and reduced expression of anti-apoptotic proteins such as Bcl-2 [194, 243, 257, 272, 290].

More recently, other forms of cell death such as pyroptosis have also been investigated in human patients [24] as well as rodent models of CCH [206, 312]. Autophagy has gained interest as well, having been shown to be upregulated specifically in VaD [41] and in CCH rodent models [47, 51, 126, 306].

These findings, in relation to cell death mechanisms being implicated in the pathophysiology of CCH, reinforce the concept of degeneration over the course of VCI progression, and may suggest that therapeutic interventions in cell death pathways may prove effective in curbing the pathological progression of VCI.

Blood–brain barrier dysfunction

The blood–brain barrier is a selectively permeable barrier that separates the circulating blood from the parenchymal tissue. The endothelial cells of the BBB are characterized by expression of tight junction proteins between adjacent cells, reduced rate of transcytosis and other transcellular movement across the barrier into or out of the brain. This property of blood–brain barrier establishes a finely tuned microenvironment for the brain by maintaining homeostasis and defending against pathogenic infections. In CCH, increased blood–brain barrier permeability has been observed [40, 191, 214, 277, 292], and is associated with neuronal loss and white matter degeneration during disease progression [271]. Blood–brain barrier damage can be induced through increased excitotoxicity, inflammation and oxidative stress, which can contribute to further brain injury via mechanisms such as increased leukocyte infiltration.

Excitotoxicity causes a persistent activation of endothelial cells causing cell death and uncontrolled movement of substance across the blood–brain barrier [6, 59]. Separately, high Ca2+ levels in the cytosol of the endothelial cells can also lead to activation of cell death mechanisms and the increased propagation of Ca2+ levels through the intracellular sources of Ca2+ such as mitochondria, and endoplasmic reticulum [92] can relocate the endothelial tight junction proteins [32]. Presence of proinflammatory cytokines can directly damage the blood–brain barrier, reducing its integrity by inducing endocytosis of the tight junction proteins thereby weakening the tight junction assembly [96, 307]. The internalised tight junction proteins are directed to lysosomal degradation, leading to long-term blood–brain barrier dysfunction [250, 279]. Reactive oxygen species have also been implicated in the progression of VaD and could possibly contribute to the breakdown of the blood–brain barrier [211]. Increased reactive oxygen species in endothelial cells downregulates epithelial cadherin levels [2] and bioavailability of nitric oxide, leading to endothelial and blood–brain barrier dysfunction [55, 89].

Blood–brain barrier dysfunction, oxidative stress [112] and inflammation induce matrix metalloproteinases (MMPs)-mediated proteolytic degradation of the extracellular matrix [63, 223, 250]. In both VaD and experimental CCH, increased levels of gelatinases (MMP-2 and MMP-9) have been reported [46, 223], which are associated with the degradation of basement membrane and tight junction proteins of the blood–brain barrier [160, 276]. Blood–brain barrier damage also increased the size of the perivascular spaces leading to cellular damage of pericytes within, a common observation in VaD. Association of pericyte damage with CCH, white matter damage, neuronal loss and cognitive impairment is evident [185] although a direct link between pericytes to VaD has yet to be demonstrated.

The blood–brain barrier regulates immune cell infiltration by maintaining low levels of leukocyte adhesion molecules on endothelial cells with inhibitory effects derived from pericytes [289]. In VaD, inflammation increases expression of adhesion molecules and chemokines such as intercellular adhesion molecule-1 (ICAM-1) and vascular adhesion molecule (VCAM) in endothelial cells [259], facilitating leukocyte infiltration [165, 171]. Upon crossing the damaged blood–brain barrier, activated leukocytes cause irreversible damage to the blood–brain barrier and contribute to further release of pro-inflammatory cytokines and reactive oxygen species, which forms a vicious feedback loop of activating endothelia [50]. Although increased ICAM levels have been reported in post-mortem studies of VaD patients [180], evidence showing specific temporal dynamics of leukocyte movement into the brain is still lacking. Overall, the establishment of endothelial cell activation upon CCH not just damages the blood–brain barrier, it also causes a reduction in the resting cerebral blood flow, and thus further contributes to a hypoperfused state within the brain. The biggest challenge here is the myriad aspects of blood–brain barrier damage and its downstream mechanisms in the context of VCI and other neuropathologies that remain unknown.

White matter lesions

One of the major pathological hallmarks of VCI is the formation of white matter lesions (WMLs) [18, 286]. The white matter functions to connect and preserve neural circuit signaling, thus implying the clinical importance of WMLs as markers of brain dysfunction due to cerebral vessel disease. Pathologically WMLs represent processes ranging from demyelination, astrogliosis, axonal loss and venular damage. These are in turn a consequence of the combined effects of increased oxidative stress and inflammation in the brain induced by CCH and blood–brain barrier breakdown [150, 163]. Our group has reported that disruption to the structural integrity of white matter can cause cognitive dysfunction [107, 123]. Anatomically, the white matter region comprises of numerous nerve fiber tracts that are surrounded by myelin. During disease progression, demyelination may occur due to various reasons. Excitotoxicity, oxidative stress and inflammation lead to oligodendrocyte damage through the loss of cellular function, mitochondrial dysfunction and production of pro-apoptotic signaling proteins, eventually contributing to their death and white matter injury [179, 255], and causing primary or secondary myelin destruction in white matter regions [178].

There is limited evidence regarding remyelination at the site of white matter injury following CCH. Remyelination is uncommon during CCH as the chronic hypoxic and pro-oxidative states block the ability of oligodendrocyte progenitor cells from being able to differentiate into newly matured oligodendrocytes [86, 91], a process further impeded by surrounding damaged endothelial cells and scar-formation during astrogliosis [10]. The age-dependent Wnt signaling pathway, which plays a role in the oligodendrocyte progenitor cells differentiation, is also compromised in VCI disease states leading to further remyelination dysfunction [130]. Nevertheless, despite being vulnerable to vascular injury, evidence showed restoration of oligodendrocyte progenitor cells and oligodendrocytes after prolonged CCH (i.e. 1 month of bilateral common carotid artery stenosis), suggesting their potential regeneration ability [181]. Studies suggested that oligodendrogenesis and regeneration are facilitated by reactive astrocytes, which secrete trophic factors such as brain-derived neurotrophic factor in response to white matter injury [169, 183, 184].

Epigenetic and genetic mechanisms

While VCI progression is mostly sporadic in nature, some forms of VCI are known to be influenced by the interplay of genetics and epigenetics. With technological advancements, researchers have improved access to diagnostic tools which carry out high throughput genomic-based investigations. The following section highlights recent research in the genetic factors of VCI as well as emerging interest in the epigenetics of VCI progression.

Epigenetics

Epigenetics refers to the alteration of gene expression without altering corresponding DNA sequences [170]. Epigenetic mechanisms are driven primarily by environmental stimuli including stress, diet and other behavioural factors. Given that most of the risk factors of VaD are associated with lifestyle-associated conditions such as hypertension and diabetes mellitus, the role of epigenetics seems to be critical in explaining the pathophysiology of the disease [203]. In fact, there are several lines of evidence for epigenetic contribution in the pathophysiology of dementia in general. These include studies where DNA methylation and hydroxymethylation were observed to be significantly reduced in the hippocampus, entorhinal cortex, cerebellum, and prefrontal cortex of AD patients compared to healthy controls [170]. Epigenetic modifications in AD neuropathology have been increasingly studied with the findings also implicated to other neurodegenerative diseases [79]. While there is limited evidence for the role of epigenetics specifically in VCI or CCH [203, 237, 299], this is likely due to the nascent nature of this topic, pointing to the need for further studies. Nevertheless, with epigenetic changes seen as drivers of pathological conditions, they may be regarded as biomarkers for early disease detection [199]. As such, further study of epigenetics would provide insights into VCI and perhaps aid in the stratification within VCI.

Genetic mutations

Certain forms of VCI onset and progression are known to have a familial component though the majority are sporadic cases [173]. Monogenic influences of tissue responses to VCI include NOTCH3 mutations causing cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), a rare form of cerebrovascular disease. The Notch pathway is important in the regulation of cell fate [121]. In particular, the Notch 3 receptor-mediated pathway is involved in the vascular smooth muscle survival [173]. In CADASIL, the mutation of the NOTCH3 gene occurs within the epidermal growth factor—like repeat domains in the N terminal of the receptor. Brains of CADASIL patients manifest an aberrant oligomerization of mutant Notch 3 proteins, leading to altered protein–protein interactions [165]. The pathophysiology of CADASIL still remains unknown, but the NF-κB pathway has been reported to play an essential role in the inflammatory responses in the CADASIL-associated angiopathy. NF-κB promotes the expression of genes coding for cytokines that leads to an amplified vascular inflammation level and hence vascular dysfunction [149]. Notch 3 misfolding phenomenon can cause an increase in free radical production in the brain, although the levels produced may not be directly pathogenic [39].

There are also overlapping genes with AD which are known to be involved in the VCI pathogenesis, namely the presenilins, the amyloid precursor protein (APP), and the apolipoprotein E (APOE) [158, 220]. It has been reported that the presence of even a single allele of the APOE4 variant could be a potential risk factor for progression of VCI [220], thus providing evidence for a commensal interaction between AD and other CVD conditions.

Unravelling the potential of early detection and intervention strategies

Currently, the treatment options for VCI remain sparse. Understanding the underlying pathophysiology of VCI through CCH provides critical insights to the discovery of biomarkers and targets for disease-modifying treatments.

The identification of specific biomarkers for VCI will be critical for more specific and sensitive diagnosis. These biomarkers may allow for early detection of VCI in at-risk patients. While the diagnostic criteria for VCI is based primarily on neuroimaging, blood-based biomarkers are nevertheless useful as surrogate disease indicators. Many blood and cerebrospinal fluid biomarkers have been identified over the years. Several proinflammatory molecule, such as C-reactive protein, IL-1α and IL-6, have been proposed as potential biomarkers [57, 133, 288]. Given that the increase in plasma level of inflammatory proteins precedes cognitive impairment in VCI, the identification of proinflammatory proteins in early stages of the disease not only offers prognostic advantage but also possible therapeutic intervention [77]. Other than inflammation, the classic marker for blood–brain barrier dysfunction, matrix metalloproteinases, has also been reported multiple times in VCI patients and found to be an early biomarker for cognitive dysfunction [73, 78, 191]. While evidence for oxidative stress in VCI patients are limited, it has been shown that oxidative stress is increased in mild cognitive impairment and AD [44, 210]. Our team has contributed to the field in establishing several possible blood biomarkers for white matter hyperintensities and microinfarcts in clinical cohorts of VCI patients such as serum hepatocyte growth factors, IL-8 and growth differentiation factor-15 [45, 106, 122, 314, 315].

Beyond specific aspects of pathophysiology, several multi-purpose therapeutic interventions have been proposed. The basis of these therapeutic interventions is built upon the evidence that cerebrovascular injury is not always progressive but may instead be reversible. For instance, white matter hyperintensities which are indicative of white matter lesions may regress and be amendable to treatments [87]. Therefore, to the extent that cerebrovascular disease such as white matter hyperintensities is related to CCH and can contribute to the risk of VCI development, markers which allow for the early identification of these lesions may enable early mitigation of cerebrovascular disease and in turn, VCI development. In a similar vein, a deep understanding of the molecular underpinnings of CCH which are relevant to cerebrovascular disease allows for the identification of potential treatment markers as well as drug targets. In the latter case, this then facilitates a potential for development of disease-modifying treatments.

Given that chronic diseases such as VCI are linked to diet and lifestyle factors, interventions at this stage are important for managing the disease. Recent studies have found association of VCI with dietary habits, shedding light on using intermittent fasting as a possible treatment for VCI [201, 280]. Intermittent fasting has been shown to improve cognitive ability, neurotropic factor production, synaptic plasticity, mitochondrial biogenesis, and has also been shown to ameliorate vascular pathology and cognitive impairment in rodent VCI models [11, 85, 110, 213, 237, 273]. Additional systemic beneficial effects of intermittent fasting include attenuation of inflammation, oxidative stress, mitochondrial dysfunction and DNA damage [65]. Separately, in clinical studies on VaD, increasing physical activity has been suggested to reduce the risk of dementia manifestation, albeit not with entirely consistent results due to lack of standardized methods [1, 71, 249, 285].

These approaches are not only potentially disease-modifying at the pathophysiological level, but they also serve as a preventive strategy to mitigate an at-risk patient’s risk of disease. More studies are required for a more robust conclusion in order to delineate the role of intermittent fasting and exercise in VCI clearly [124, 156, 157, 235]. Notably, our team have been investigating the effect of multiple lifestyle interventions on the prevention of cognitive decline under the Singapore Geriatric Intervention Study to Reduce Cognitive Decline and Physical Frailty (SINGER) [52], which is an adaptation of the pioneering Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER) [151]. Lifestyle interventions provide a promising potential in managing VCI and reframing the public health perspective of the disease.

Summary and future directions

VCI is now a widely accepted term introduced to embody the entire spectrum of vascular-related cognitive alterations or cerebrovascular disease-related burdens that can manifest into cognitive impairments [100]. Cognitive deficits associated with VCI include slower mental processing and impaired executive functioning such as poor planning, poor judgement and poor decision-making. Non-cognitive behavioural manifestations include apathy, anxiety and even depression are also common. VCI has been gaining interest in the field as it is potentially preventable, prior to reaching the end-stage dementia [132, 294]. Given such emphasis on early detection and diagnosis in the field, there is a need to better understand the pathophysiology of VCI. As reviewed above, current experimental evidence indicates that a chronic state of hypoperfusion in the brain drives the various pathophysiological mechanisms and structural changes in the brain. CCH therefore holds promise in shedding some light on the molecular and mechanistic underpinnings of VCI.

As reviewed above, there are several common mechanisms that occur during the progression of CCH-induced injury such as energy imbalance, oxidative stress, endoplasmic reticulum stress, mitochondrial dysfunction and inflammation (Fig. 2). These mechanisms drive the downstream structural neuropathological changes in the brain including glial activation, cell death activation, blood–brain barrier breakdown and white matter lesion formation. The pathological features begin from a hypoperfused state and can coexist and interact to adversely influence cognitive function as reported in animal models [25, 27, 43, 166]. This suggests that the effects of CCH on cognition are mediated by mechanistic drivers and structural changes in the brain. Indeed, CCH may be the earliest, insidious indicator of VCI, while brain atrophy and white matter lesions may occur downstream from CCH as more dynamic and detectable changes.

It is exciting to witness the field of VCI rapidly expanding and moving towards sharper definitions and deeper insights into underlying mechanisms. The heterogeneity of the disease is widely recognized to be due to the complex interactions between vascular injuries and risk factors that are involved prior to, and during disease manifestation. Across these subtypes, variations also exist at the clinical, neuroimaging, and pathological levels. Yet, a strong argument may be made that all subtypes of VCI include a CCH state, which we believe to be the main driver for subsequent pathological progression. Prolonged cerebral hypoperfusion may therefore serve as the transition from the at-risk state to the VCI state. The observational and experimental evidence from CCH models presented in this review help reinforce the importance of CCH as a critical feature in our efforts to unravel the underlying molecular mechanisms of VCI. Further identification of specific biomarkers of CCH may provide the rationale for the evaluation of these markers in the clinic which can bring us closer to detecting VCI at an early stage as well as introduce treatment options which may delay disease onset or slow disease progression.

References

  1. Aarsland D, Saeedzadeh-Sardahaee F, Anderssen S, Ballard C (2010) Is physical activity a potential preventive factor for vascular dementia? A systematic review. Aging Ment Health. https://doi.org/10.1080/13607860903586136

    Article  PubMed  Google Scholar 

  2. Abbruscato T, Davis T (1999) Protein expression of brain endothelial cell E-cadherin after hypoxia/aglycemia: influence of astrocyte contact. Brain Res 842:277–286. https://doi.org/10.1016/S0006-8993(99)01778-3

    Article  CAS  PubMed  Google Scholar 

  3. Abraham JM, Cho L (2010) The homocysteine hypothesis: still relevant to the prevention and treatment of cardiovascular disease? Cleve Clin J Med 77:911–918. https://doi.org/10.3949/ccjm.77a.10036

    Article  PubMed  Google Scholar 

  4. Aevarsson Ó, Svanborg A, Skoog I (1998) Seven-year survival rate after age 85 years: relation to Alzheimer disease and vascular dementia. Arch Neurol 55:1226–1232. https://doi.org/10.1001/ARCHNEUR.55.9.1226

    Article  CAS  PubMed  Google Scholar 

  5. Agüero-Torres H, Fratiglioni L, Guo Z, Viitanen M, Winblad B (1999) Mortality from dementia in advanced age: a 5-year follow-up study of incident dementia cases. J Clin Epidemiol 52:737–743. https://doi.org/10.1016/S0895-4356(99)00067-0

    Article  PubMed  Google Scholar 

  6. András IE, Deli MA, Veszelka S, Hayashi K, Hennig B, Toborek M (2007) The NMDA and AMPA/KA receptors are involved in glutamate-induced alterations of occludin expression and phosphorylation in brain endothelial cells. J Cereb Blood Flow Metab 27:1431–1443. https://doi.org/10.1038/SJ.JCBFM.9600445

    Article  PubMed  Google Scholar 

  7. Anjum I, Fayyaz M, Wajid A, Sohail W, Ali A (2018) Does obesity increase the risk of dementia: a literature review. Cureus 10:e2660. https://doi.org/10.7759/cureus.2660

    Article  PubMed  PubMed Central  Google Scholar 

  8. Anstey KJ, von Sanden C, Salim A, O’Kearney R (2007) Smoking as a risk factor for dementia and cognitive decline: a meta-analysis of prospective studies. Am J Epidemiol 166:367–378. https://doi.org/10.1093/aje/kwm116

    Article  PubMed  Google Scholar 

  9. Appleton JP, Scutt P, Sprigg N, Bath PM (2017) Hypercholesterolaemia and vascular dementia. Clin Sci 131:1561–1578. https://doi.org/10.1042/cs20160382

    Article  Google Scholar 

  10. Arai K, Lo EH (2010) Astrocytes protect oligodendrocyte precursor cells via MEK/ERK and PI3K/Akt signaling. J Neurosci Res 88:758–763. https://doi.org/10.1002/jnr.22256

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Arguin H, Dionne IJ, Sénéchal M, Bouchard DR, Carpentier AC, Ardilouze J-L, Tremblay A, Leblanc C, Brochu M (2012) Short- and long-term effects of continuous versus intermittent restrictive diet approaches on body composition and the metabolic profile in overweight and obese postmenopausal women: a pilot study. Menopause 19:870

    Article  PubMed  Google Scholar 

  12. Arnoldussen IA, Kiliaan AJ, Gustafson DR (2014) Obesity and dementia: adipokines interact with the brain. Eur Neuropsychopharmacol 24:1982–1999. https://doi.org/10.1016/j.euroneuro.2014.03.002

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Aronson MK, Ooi WL, Morgenstern H, Hafner A, Masur D, Crystal H, Frishman WH, Fisher D, Katzman R (1990) Women, myocardial infarction, and dementia in the very old. Neurology 40:1102–1106. https://doi.org/10.1212/wnl.40.7.1102

    Article  CAS  PubMed  Google Scholar 

  14. Arundine M, Tymianski M (2003) Molecular mechanisms of calcium-dependent neurodegeneration in excitotoxicity. Cell Calcium 34:325–337. https://doi.org/10.1016/S0143-4160(03)00141-6

    Article  CAS  PubMed  Google Scholar 

  15. Babcock AA, Kuziel WA, Rivest S, Owens T (2003) Chemokine expression by glial cells directs leukocytes to sites of axonal injury in the CNS. J Neurosci 23:7922–7930. https://doi.org/10.1523/JNEUROSCI.23-21-07922.2003

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Baik SH, Selvaraji S, Fann DY, Poh L, Jo DG, Herr DR, Zhang SR, Kim HA, Silva M, Lai MKP et al (2021) Hippocampal transcriptome profiling reveals common disease pathways in chronic hypoperfusion and aging. Aging (Albany NY) 13:14651–14674. https://doi.org/10.18632/aging.203123

    Article  CAS  PubMed  Google Scholar 

  17. Bakhru A, Erlinger TP (2005) Smoking cessation and cardiovascular disease risk factors: results from the Third National Health and Nutrition Examination Survey. PLoS Med 2:e160. https://doi.org/10.1371/journal.pmed.0020160

    Article  PubMed  PubMed Central  Google Scholar 

  18. Bakker SLM, de Leeuw FE, de Groot JC, Hofman A, Koudstaal PJ, Breteler MMB (1999) Cerebral vasomotor reactivity and cerebral white matter lesions in the elderly. Neurology 52:578–578. https://doi.org/10.1212/WNL.52.3.578

    Article  CAS  PubMed  Google Scholar 

  19. Barić D (2014) Why pulsatility still matters: a review of current knowledge. Croat Med J 55:609–620. https://doi.org/10.3325/cmj.2014.55.609

    Article  PubMed  PubMed Central  Google Scholar 

  20. Barna BP, Pettay J, Barnett GH, Zhou P, Iwasaki K, Estes ML (1994) Regulation of monocyte chemoattractant protein-1 expression in adult human non-neoplastic astrocytes is sensitive to tumor necrosis factor (TNF) or antibody to the 55-kDa TNF receptor. J Neuroimmunol 50:101–107. https://doi.org/10.1016/0165-5728(94)90220-8

    Article  CAS  PubMed  Google Scholar 

  21. Barnes DE, Yaffe K, Byers AL, McCormick M, Schaefer C, Whitmer RA (2012) Midlife vs late-life depressive symptoms and risk of dementia: differential effects for Alzheimer disease and vascular dementia. Arch Gen Psychiatry 69:493–498. https://doi.org/10.1001/archgenpsychiatry.2011.1481

    Article  PubMed  PubMed Central  Google Scholar 

  22. Barreras A, Gurk-Turner C (2003) Angiotensin II receptor blockers. In: Baylor University Medical Center Proceedings, vol 16, pp 123–126. https://doi.org/10.1080/08998280.2003.11927893

  23. Barreto G, White RE, Ouyang Y, Xu L, Giffard RG (2011) Astrocytes: targets for neuroprotection in stroke. Cent Nerv Syst Agents Med Chem 11:164–173. https://doi.org/10.2174/187152411796011303

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Belkhelfa M, Beder N, Mouhoub D, Amri M, Hayet R, Nabila T, Bakhti S, Laimouche S, Azzouz D, Belhadj R et al (2018) The involvement of neuroinflammation and necroptosis in the hippocampus during vascular dementia. J Neuroimmunol. https://doi.org/10.1016/j.jneuroim.2018.04.004

    Article  PubMed  Google Scholar 

  25. Ben Ari H, Lifschytz T, Wolf G, Rigbi A, Blumenfeld-Katzir T, Kreisel Merzel T, Koroukhov N, Lotan A, Lerer B (2019) White matter lesions, cerebral inflammation and cognitive function in a mouse model of cerebral hypoperfusion. Brain Res. https://doi.org/10.1016/j.brainres.2019.01.017

    Article  PubMed  Google Scholar 

  26. Bennett S, Grant MM, Aldred S (2009) Oxidative stress in vascular dementia and Alzheimer’s disease: a common pathology. J Alzheimers Dis 17:245–257. https://doi.org/10.3233/JAD-2009-1041

    Article  CAS  PubMed  Google Scholar 

  27. Bennett SA, Tenniswood M, Chen JH, Davidson CM, Keyes MT, Fortin T, Pappas BA (1998) Chronic cerebral hypoperfusion elicits neuronal apoptosis and behavioral impairment. NeuroReport 9:161–166. https://doi.org/10.1097/00001756-199801050-00033

    Article  CAS  PubMed  Google Scholar 

  28. Bonifati DM, Kishore U (2007) Role of complement in neurodegeneration and neuroinflammation. Mol Immunol 44:999–1010. https://doi.org/10.1016/j.molimm.2006.03.007

    Article  CAS  PubMed  Google Scholar 

  29. Bowling AC, Mutisya EM, Walker LC, Price DL, Cork LC, Beal MH (1993) Age-dependent impairment of mitochondrial function in primate brain. J Neurochem 60:1964–1967. https://doi.org/10.1111/j.1471-4159.1993.tb13430.x

    Article  CAS  PubMed  Google Scholar 

  30. Breteler MM, Claus JJ, Grobbee DE, Hofman A (1994) Cardiovascular disease and distribution of cognitive function in elderly people: the Rotterdam study. BMJ 308:1604–1608. https://doi.org/10.1136/bmj.308.6944.1604

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Brown MD, Wallace DC (1994) Molecular basis of mitochondrial DNA disease. J Bioenerg Biomembr 26:273–289. https://doi.org/10.1007/BF00763099

    Article  CAS  PubMed  Google Scholar 

  32. Brown RC, Davis TP (2002) Calcium modulation of adherens and tight junction function. Stroke 33:1706–1711. https://doi.org/10.1161/01.STR.0000016405.06729.83

    Article  CAS  PubMed  Google Scholar 

  33. Brown WR, Moody DM, Thore CR, Challa VR (2000) Apoptosis in leukoaraiosis. Am J Neuroradiol 21:79–82

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Brown WR, Thore CR (2011) Review: cerebral microvascular pathology in ageing and neurodegeneration. Neuropathol Appl Neurobiol 37:56–74. https://doi.org/10.1111/j.1365-2990.2010.01139.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Brun A (1994) Pathology and pathophysiology of cerebrovascular dementia: pure subgroups of obstructive and hypoperfusive etiology. Dement Geriatr Cogn Disord 5:145–147. https://doi.org/10.1159/000106712

    Article  CAS  Google Scholar 

  36. Bunch TJ, Weiss JP, Crandall BG, May HT, Bair TL, Osborn JS, Anderson JL, Muhlestein JB, Horne BD, Lappe DL et al (2010) Atrial fibrillation is independently associated with senile, vascular, and Alzheimer’s dementia. Heart Rhythm 7:433–437. https://doi.org/10.1016/j.hrthm.2009.12.004

    Article  PubMed  Google Scholar 

  37. Byers AL, Yaffe K (2011) Depression and risk of developing dementia. Nat Rev Neurol 7:323–331. https://doi.org/10.1038/nrneurol.2011.60

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Camacho A, Massieu L (2006) Role of glutamate transporters in the clearance and release of glutamate during Ischemia and its relation to neuronal death. Arch Med Res 37:11–18. https://doi.org/10.1016/j.arcmed.2005.05.014

    Article  CAS  PubMed  Google Scholar 

  39. Campolo J, De Maria R, Mariotti C, Tomasello C, Parolini M, Frontali M, Inzitari D, Valenti R, Federico A, Taroni F et al (2013) Is the oxidant/antioxidant status altered in CADASIL patients? PLoS ONE 8:e67077–e67077. https://doi.org/10.1371/journal.pone.0067077

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Candelario-Jalil E, Thompson J, Taheri S, Grossetete M, Adair JC, Edmonds E, Prestopnik J, Wills J, Rosenberg GA (2011) Matrix metalloproteinases are associated with increased blood–brain barrier opening in vascular cognitive impairment. Stroke 42:1345–1350. https://doi.org/10.1161/STROKEAHA.110.600825

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Castellazzi M, Patergnani S, Donadio M, Giorgi C, Bonora M, Bosi C, Brombo G, Pugliatti M, Seripa D, Zuliani G et al (2019) Autophagy and mitophagy biomarkers are reduced in sera of patients with Alzheimer’s disease and mild cognitive impairment. Sci Rep 9:20009–20009. https://doi.org/10.1038/s41598-019-56614-5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Catindig J-AS, Venketasubramanian N, Ikram MK, Chen C (2012) Epidemiology of dementia in Asia: insights on prevalence, trends and novel risk factors. J Neurol Sci 321:11–16

    Article  PubMed  Google Scholar 

  43. Cechetti F, Pagnussat AS, Worm PV, Elsner VR, Ben J, da Costa MS, Mestriner R, Weis SN, Netto CA (2012) Chronic brain hypoperfusion causes early glial activation and neuronal death, and subsequent long-term memory impairment. Brain Res Bull 87:109–116. https://doi.org/10.1016/j.brainresbull.2011.10.006

    Article  CAS  PubMed  Google Scholar 

  44. Cervellati C, Romani A, Seripa D, Cremonini E, Bosi C, Magon S, Bergamini CM, Valacchi G, Pilotto A, Zuliani G (2014) Systemic oxidative stress and conversion to dementia of elderly patients with mild cognitive impairment. Biomed Res Int 2014:309507–309507. https://doi.org/10.1155/2014/309507

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Chai YL, Hilal S, Chong JR, Ng YX, Liew OW, Xu X, Ikram MK, Venketasubramanian N, Richards AM, Lai MKP et al (2016) Growth differentiation factor-15 and white matter hyperintensities in cognitive impairment and dementia. Medicine 95:e4566. https://doi.org/10.1097/MD.0000000000004566

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Chai YL, Rajeev V, Poh L, Selvaraji S, Hilal S, Chen CP, Jo DG, Koo EH, Arumugam TV, Lai MK (2022) Chronic cerebral hypoperfusion alters the CypA-EMMPRIN-gelatinase pathway: implications for vascular dementia. J Cereb Blood Flow Metab. https://doi.org/10.1177/0271678x221146401

    Article  PubMed  Google Scholar 

  47. Che H, Yan Y, Kang X-H, Guo F, Yan M-L, Liu H-L, Hou X, Liu T, Zong D-K, Sun L-L et al (2017) MicroRNA-27a promotes inefficient lysosomal clearance in the hippocampi of rats following chronic brain hypoperfusion. Mol Neurobiol 54:2595–2610. https://doi.org/10.1007/s12035-016-9856-8

    Article  CAS  PubMed  Google Scholar 

  48. Chen C, Homma A, Mok VCT, Krishnamoorthy E, Alladi S, Meguro K, Abe K, Dominguez J, Marasigan S, Kandiah N (2016) Alzheimer’s disease with cerebrovascular disease: current status in the Asia–Pacific region. J Intern Med 280:359–374

    Article  CAS  PubMed  Google Scholar 

  49. Chen CPLH (2004) Transcultural expression of subcortical vascular disease. J Neurol Sci 226:45–47. https://doi.org/10.1016/j.jns.2004.09.010

    Article  PubMed  Google Scholar 

  50. Chen L, Deng H, Cui H, Fang J, Zuo Z, Deng J, Li Y, Wang X, Zhao L (2017) Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 9:7204–7218. https://doi.org/10.18632/oncotarget.23208

    Article  PubMed  PubMed Central  Google Scholar 

  51. Chen Y, Guo Z, Peng X, Xie W, Chen L, Tan Z (2018) Nimodipine represses AMPK phosphorylation and excessive autophagy after chronic cerebral hypoperfusion in rats. Brain Res Bull 140:88–96. https://doi.org/10.1016/j.brainresbull.2018.03.019

    Article  CAS  PubMed  Google Scholar 

  52. Chew KA, Xu X, Siongco P, Villaraza S, Phua AKS, Wong ZX, Chung CY, Tang N, Chew E, Henry CJ et al (2021) SINgapore GERiatric intervention study to reduce physical frailty and cognitive decline (SINGER)-pilot: a feasibility study. Alzheimers Dement (NY) 7:e12141. https://doi.org/10.1002/trc2.12141

    Article  Google Scholar 

  53. Choi DH, Lee KH, Kim JH, Seo JH, Kim HY, Shin CY, Han JS, Han SH, Kim YS, Lee J (2014) NADPH oxidase 1, a novel molecular source of ROS in hippocampal neuronal death in vascular dementia. Antioxid Redox Signal 21:533–550. https://doi.org/10.1089/ars.2012.5129

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Choi JC (2015) Genetics of cerebral small vessel disease. J Stroke 17:7–16. https://doi.org/10.5853/jos.2015.17.1.7

    Article  PubMed  PubMed Central  Google Scholar 

  55. Chrissobolis S, Banfi B, Sobey CG, Faraci FM (2012) Role of Nox isoforms in angiotensin II-induced oxidative stress and endothelial dysfunction in brain. J Appl Physiol 113:184–191. https://doi.org/10.1152/JAPPLPHYSIOL.00455.2012

    Article  CAS  PubMed  Google Scholar 

  56. Ciacciarelli A, Sette G, Giubilei F, Orzi F (2020) Chronic cerebral hypoperfusion: an undefined, relevant entity. J Clin Neurosci. https://doi.org/10.1016/j.jocn.2020.01.026

    Article  PubMed  Google Scholar 

  57. Cipollini V, Troili F, Giubilei F (2019) Emerging biomarkers in vascular cognitive impairment and dementia: from pathophysiological pathways to clinical application. Int J Mol Sci 20:2812–2812. https://doi.org/10.3390/ijms20112812

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Colcombe S, Kramer AF (2003) Fitness effects on the cognitive function of older adults: a meta-analytic study. Psychol Sci 14:125–130. https://doi.org/10.1111/1467-9280.t01-1-01430

    Article  PubMed  Google Scholar 

  59. Collard CD, Park KA, Montalto MC, Alapati S, Buras JA, Stahl GL, Colgan SP (2002) Neutrophil-derived Glutamate regulates vascular endothelial barrier function. J Biol Chem 277:14801–14811. https://doi.org/10.1074/JBC.M110557200

    Article  CAS  PubMed  Google Scholar 

  60. Corrada MM, Brookmeyer R, Berlau D, Paganini-Hill A, Kawas CH (2008) Prevalence of dementia after age 90: results from the 90+ study. Neurology 71:337–343. https://doi.org/10.1212/01.wnl.0000310773.65918.cd

    Article  CAS  PubMed  Google Scholar 

  61. Corraini P, Henderson VW, Ording AG, Pedersen L, Horváth-Puhó E, Sørensen HT (2017) Long-term risk of dementia among survivors of ischemic or hemorrhagic stroke. Stroke 48:180–186. https://doi.org/10.1161/strokeaha.116.015242

    Article  CAS  PubMed  Google Scholar 

  62. Courties G, Moskowitz MA, Nahrendorf M (2014) The innate immune system after ischemic injury: lessons to be learned from the heart and brain. JAMA Neurol 71:233–236. https://doi.org/10.1001/jamaneurol.2013.5026

    Article  PubMed  PubMed Central  Google Scholar 

  63. Daneman R, Prat A (2015) The blood–brain barrier. Cold Spring Harb Perspect Biol 7:a020412. https://doi.org/10.1101/CSHPERSPECT.A020412

    Article  PubMed  Google Scholar 

  64. Datta A, Qian J, Chong R, Kalaria RN, Francis P, Lai MKP, Chen CP, Sze SK (2014) Novel pathophysiological markers are revealed by iTRAQ-based quantitative clinical proteomics approach in vascular dementia. J Proteom 99:54–67. https://doi.org/10.1016/j.jprot.2014.01.011

    Article  CAS  Google Scholar 

  65. de Cabo R, Mattson MP (2019) Effects of intermittent fasting on health, aging, and disease. N Engl J Med 381:2541–2551. https://doi.org/10.1056/NEJMra1905136

    Article  PubMed  Google Scholar 

  66. de la Torre JC (2012) Cardiovascular risk factors promote brain hypoperfusion leading to cognitive decline and dementia. Cardiovasc Psychiatry Neurol 2012:367516. https://doi.org/10.1155/2012/367516

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. De Silva TM, Faraci FM (2016) Microvascular dysfunction and cognitive impairment. Cell Mol Neurobiol 36:241–258. https://doi.org/10.1007/s10571-015-0308-1

    Article  PubMed  PubMed Central  Google Scholar 

  68. De Vivo DC (1993) The expanding clinical spectrum of mitochondrial diseases. Brain Dev 15:1–22. https://doi.org/10.1016/0387-7604(93)90002-P

    Article  PubMed  Google Scholar 

  69. Del Ser T, Hachinski V, Merskey H, Munoz DG (1999) An autopsy-verified study of the effect of education on degenerative dementia. Brain 122(Pt 12):2309–2319. https://doi.org/10.1093/brain/122.12.2309

    Article  PubMed  Google Scholar 

  70. Ding Q, Dimayuga E, Keller JN (2006) Proteasome regulation of oxidative stress in aging and age-related diseases of the CNS. Antioxid Redox Signal 8:163–172. https://doi.org/10.1089/ars.2006.8.163

    Article  CAS  PubMed  Google Scholar 

  71. Dong J, Zhao J, Lin Y, Liang H, He X, Zheng X, Sui M, Zhuang Z, Yan T (2018) Exercise improves recognition memory and synaptic plasticity in the prefrontal cortex for rats modelling vascular dementia. Neurol Res 40:68–77. https://doi.org/10.1080/01616412.2017.1398389

    Article  PubMed  Google Scholar 

  72. Du J, Ma M, Zhao Q, Fang L, Chang J, Wang Y, Fei R, Song X (2012) Mitochondrial bioenergetic deficits in the hippocampus of rats with chronic Ischemia-induced vascular dementia. Neuroscience. https://doi.org/10.1016/j.neuroscience.2012.11.062

    Article  PubMed  PubMed Central  Google Scholar 

  73. Duits FH, Hernandez-Guillamon M, Montaner J, Goos JDC, Montañola A, Wattjes MP, Barkhof F, Scheltens P, Teunissen CE, van der Flier WM (2015) Matrix metalloproteinases in Alzheimer’s disease and concurrent cerebral microbleeds. J Alzheimer’s Dis 48:711–720. https://doi.org/10.3233/JAD-143186

    Article  CAS  Google Scholar 

  74. Duncombe J, Kitamura A, Hase Y, Ihara M, Kalaria Raj N, Horsburgh K (2017) Chronic cerebral hypoperfusion: a key mechanism leading to vascular cognitive impairment and dementia. Closing the translational gap between rodent models and human vascular cognitive impairment and dementia. Clin Sci 131:2451–2468. https://doi.org/10.1042/CS20160727

    Article  CAS  Google Scholar 

  75. Duron E, Hanon O (2008) Vascular risk factors, cognitive decline, and dementia. Vasc Health Risk Manag 4:363–381. https://doi.org/10.2147/vhrm.s1839

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Enciu A-M, Constantinescu SN, Popescu LM, Mureşanu DF, Popescu BO (2011) Neurobiology of vascular dementia. J Aging Res 2011:401604. https://doi.org/10.4061/2011/401604

    Article  PubMed  PubMed Central  Google Scholar 

  77. Engelhart MJ, Geerlings MI, Meijer J, Kiliaan A, Ruitenberg A, van Swieten JC, Stijnen T, Hofman A, Witteman JCM, Breteler MMB (2004) Inflammatory proteins in plasma and the risk of dementia: the rotterdam study. Arch Neurol 61:668–672. https://doi.org/10.1001/archneur.61.5.668

    Article  PubMed  Google Scholar 

  78. Erhardt EB, Pesko JC, Prestopnik J, Thompson J, Caprihan A, Rosenberg GA (2018) Biomarkers identify the Binswanger type of vascular cognitive impairment. J Cereb Blood Flow Metab 39:1602–1612. https://doi.org/10.1177/0271678X18762655

    Article  Google Scholar 

  79. Esposito M, Sherr GL (2019) Epigenetic modifications in Alzheimer’s neuropathology and therapeutics. Front Neurosci 13:476–476. https://doi.org/10.3389/fnins.2019.00476

    Article  PubMed  PubMed Central  Google Scholar 

  80. Famulari AL, Marschoff ER, Llesuy SF, Kohan S, Serra JA, Domínguez RO, Repetto MG, Reides CG, de Lustig ES (1996) The antioxidant enzymatic blood profile in Alzheimer’s and vascular diseases. Their association and a possible assay to differentiate demented subjects and controls. J Neurol Sci 141:69–78

    Article  CAS  PubMed  Google Scholar 

  81. Fan Y, Ou X, Wang W, Tian X, Yan S, Hu N, Zhang X, Xing W (2017) Identification and analysis of toll like receptor 4 (TLR4) level changes in vascular dementia patients related type 2 diabetes mellitus. Biomed Res 28:4588

    CAS  Google Scholar 

  82. Fann DY-W, Lee S-Y, Manzanero S, Chunduri P, Sobey CG, Arumugam TV (2013) Pathogenesis of acute stroke and the role of inflammasomes. Ageing Res Rev 12:941–966. https://doi.org/10.1016/j.arr.2013.09.004

    Article  CAS  PubMed  Google Scholar 

  83. Faraci F (2006) Reactive oxygen species: influence on cerebral vascular tone. J Appl Physiol 100:739–743. https://doi.org/10.1152/japplphysiol.01044.2005

    Article  CAS  PubMed  Google Scholar 

  84. Favaloro B, Allocati N, Graziano V, Di Ilio C, De Laurenzi V (2012) Role of apoptosis in disease. Aging 4:330–349. https://doi.org/10.18632/aging.100459

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Fernando HA, Zibellini J, Harris RA, Seimon RV, Sainsbury A (2019) Effect of Ramadan fasting on weight and body composition in healthy non-athlete adults: a systematic review and meta-analysis. Nutrients 11:478–478. https://doi.org/10.3390/nu11020478

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Fernando MS, Simpson JE, Matthews F, Brayne C, Lewis CE, Barber R, Kalaria RN, Forster G, Esteves F, Wharton SB et al (2006) White matter lesions in an unselected cohort of the elderly. Stroke 37:1391–1398. https://doi.org/10.1161/01.STR.0000221308.94473.14

    Article  PubMed  Google Scholar 

  87. Filley CM (2021) Cognitive dysfunction in white matter disorders: new perspectives in treatment and recovery. J Neuropsychiatry Clin Neurosci 33:349–355. https://doi.org/10.1176/appi.neuropsych.21030080

    Article  PubMed  Google Scholar 

  88. Fischer S, Wobben M, Kleinstück J, Renz D, Schaper W (2000) Effect of astroglial cells on hypoxia-induced permeability in PBMEC cells. Am J Physiol Cell Physiol 279:C935–C944. https://doi.org/10.1152/ajpcell.2000.279.4.C935

    Article  CAS  PubMed  Google Scholar 

  89. Förstermann U (2010) Nitric oxide and oxidative stress in vascular disease. Pflüg Arch Eur J Physiol 459(6):923–939. https://doi.org/10.1007/S00424-010-0808-2

    Article  Google Scholar 

  90. Freiheit EA, Hogan DB, Eliasziw M, Patten SB, Demchuk AM, Faris P, Anderson T, Galbraith D, Parboosingh JS, Ghali WA et al (2012) A dynamic view of depressive symptoms and neurocognitive change among patients with coronary artery disease. Arch Gen Psychiatry 69:244–255. https://doi.org/10.1001/archgenpsychiatry.2011.1361

    Article  PubMed  Google Scholar 

  91. French HM, Reid M, Mamontov P, Simmons RA, Grinspan JB (2009) Oxidative stress disrupts oligodendrocyte maturation. J Neurosci Res 87:3076–3087. https://doi.org/10.1002/jnr.22139

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Friedman L (2007) CALCIUM: a role for neuroprotection and sustained adaptation. Mol Interv 6:315–329. https://doi.org/10.1124/mi.6.6.5

    Article  Google Scholar 

  93. Fünfschilling U, Supplie L, Mahad D, Boretius S, Saab A, Edgar J, Brinkmann B, Kassmann C, Tzvetanova I, Möbius W et al (2012) Glycolytic oligodendrocytes maintain myelin and long-term axonal integrity. Nature 485:517–521. https://doi.org/10.1038/nature11007

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Gannon OJ, Robison LS, Custozzo AJ, Zuloaga KL (2019) Sex differences in risk factors for vascular contributions to cognitive impairment & dementia. Neurochem Int 127:38–55. https://doi.org/10.1016/j.neuint.2018.11.014

    Article  CAS  PubMed  Google Scholar 

  95. Gemma C, Vila J, Bachstetter A, Bickford PC (2007) Oxidative stress and the aging brain: from theory to prevention. CRC Press/Routledge/Taylor & Francis Group, pp 353–374

    Google Scholar 

  96. González-Mariscal L, Tapia R, Chamorro D (2008) Crosstalk of tight junction components with signaling pathways. Biochim Biophys Acta 1778:729–756. https://doi.org/10.1016/J.BBAMEM.2007.08.018

    Article  PubMed  Google Scholar 

  97. Gordon P, Flanagan P (2016) Smoking: a risk factor for vascular disease. J Vasc Nurs 34:79–86. https://doi.org/10.1016/j.jvn.2016.04.001

    Article  PubMed  Google Scholar 

  98. Gorelick PB (2004) Risk factors for vascular dementia and Alzheimer disease. Stroke 35:2620–2622. https://doi.org/10.1161/01.Str.0000143318.70292.47

    Article  PubMed  Google Scholar 

  99. Gorelick PB, Counts SE, Nyenhuis D (2016) Vascular cognitive impairment and dementia. Biochim Biophys Acta (BBA) Mol Basis Dis 1862:860–868. https://doi.org/10.1016/j.bbadis.2015.12.015

    Article  CAS  Google Scholar 

  100. Gorelick PB, Scuteri A, Black SE, Decarli C, Greenberg SM, Iadecola C, Launer LJ, Laurent S, Lopez OL, Nyenhuis D et al (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. https://doi.org/10.1161/STR.0b013e3182299496

    Article  PubMed  PubMed Central  Google Scholar 

  101. Graban A, Bednarska-Makaruk M, Bochyńska A, Lipczyńska-Łojkowska W, Ryglewicz D, Wehr H (2009) Vascular and biochemical risk factors of vascular dementia after lacunar strokes (S-VaD) and after multiinfarcts in strategic areas (M-VaD). J Neurol Sci 283:116–118. https://doi.org/10.1016/j.jns.2009.02.344

    Article  PubMed  Google Scholar 

  102. Graeber MB, Grasbon-Frodl E, Eitzen UV, Kösel S (1998) Neurodegeneration and aging: role of the second genome. J Neurosci Res 52:1–6. https://doi.org/10.1002/(SICI)1097-4547(19980401)52:1%3c1::AID-JNR1%3e3.0.CO;2-I

    Article  CAS  PubMed  Google Scholar 

  103. Gray F, Polivka M, Viswanathan A, Baudrimont M, Bousser M-G, Chabriat H (2007) Apoptosis in cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy. J Neuropathol Exp Neurol 66:597–607. https://doi.org/10.1097/nen.0b013e318093e574

    Article  PubMed  Google Scholar 

  104. Grimm A, Friedland K, Eckert A (2016) Mitochondrial dysfunction: the missing link between aging and sporadic Alzheimer’s disease. Biogerontology. https://doi.org/10.1007/s10522-015-9618-4

    Article  PubMed  Google Scholar 

  105. Grivennikov S, Karin M (2010) Dangerous liaisons: STAT3 and NF-kappaB collaboration and crosstalk in cancer. Cytokine Growth Factor Rev 21(1):11–19

    Article  CAS  PubMed  Google Scholar 

  106. Gyanwali B, Lai MKP, Lui B, Liew OW, Venketasubramanian N, Richards AM, Chen C, Hilal S (2021) Blood-based cardiac biomarkers and the risk of cognitive decline, cerebrovascular disease, and clinical events. Stroke 52:2275–2283. https://doi.org/10.1161/strokeaha.120.032571

    Article  CAS  PubMed  Google Scholar 

  107. Gyanwali B, Shaik MA, Tan BY, Venketasubramanian N, Chen C, Hilal S (2019) Risk factors for and clinical relevance of incident and progression of cerebral small vessel disease markers in an Asian memory clinic population. J Alzheimer’s Dis 67:1209–1219. https://doi.org/10.3233/JAD-180911

    Article  CAS  Google Scholar 

  108. Hachinski V, Iadecola C, Petersen RC, Breteler MM, Nyenhuis DL, Black SE, Powers WJ, DeCarli C, Merino JG, Kalaria RN et al (2006) National Institute of Neurological Disorders and Stroke-Canadian Stroke Network vascular cognitive impairment harmonization standards. Stroke 37:2220–2241. https://doi.org/10.1161/01.STR.0000237236.88823.47

    Article  PubMed  Google Scholar 

  109. Hainsworth AH, Yeo NE, Weekman EM, Wilcock DM (2016) Homocysteine, hyperhomocysteinemia and vascular contributions to cognitive impairment and dementia (VCID). Biochim Biophys Acta 1862:1008–1017. https://doi.org/10.1016/j.bbadis.2015.11.015

    Article  CAS  PubMed  Google Scholar 

  110. Halberg N, Henriksen M, Söderhamn N, Stallknecht B, Ploug T, Schjerling P, Dela F (2005) Effect of intermittent fasting and refeeding on insulin action in healthy men. J Appl Physiol 99:2128–2136. https://doi.org/10.1152/japplphysiol.00683.2005

    Article  CAS  PubMed  Google Scholar 

  111. Halling A, Berglund J (2006) Association of diagnosis of ischaemic heart disease, diabetes mellitus and heart failure with cognitive function in the elderly population. Eur J Gen Pract 12:114–119. https://doi.org/10.1080/13814780600881128

    Article  PubMed  Google Scholar 

  112. Haorah J, Ramirez SH, Schall K, Smith D, Pandya R, Persidsky Y (2007) Oxidative stress activates protein tyrosine kinase and matrix metalloproteinases leading to blood–brain barrier dysfunction. J Neurochem 101:566–576. https://doi.org/10.1111/j.1471-4159.2006.04393.x

    Article  CAS  PubMed  Google Scholar 

  113. Harter K, Levine M, Henderson SO (2015) Anticoagulation drug therapy: a review. West J Emerg Med 16:11–17. https://doi.org/10.5811/westjem.2014.12.22933

    Article  PubMed  PubMed Central  Google Scholar 

  114. Hattori Y, Enmi J, Kitamura A, Yamamoto Y, Saito S, Takahashi Y, Iguchi S, Tsuji M, Yamahara K, Nagatsuka K et al (2015) A novel mouse model of subcortical infarcts with dementia. J Neurosci 35:3915–3928. https://doi.org/10.1523/jneurosci.3970-14.2015

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Hayashi M, Luo Y, Laning J, Strieter RM, Dorf ME (1995) Production and function of monocyte chemoattractant protein-1 and other β-chemokines in murine glial cells. J Neuroimmunol 60:143–150. https://doi.org/10.1016/0165-5728(95)00064-9

    Article  CAS  PubMed  Google Scholar 

  116. Helmer C, Joly P, Letenneur L, Commenges D, Dartigues JF (2001) Mortality with dementia: results from a French prospective community-based cohort. Am J Epidemiol 154:642–648. https://doi.org/10.1093/aje/154.7.642

    Article  CAS  PubMed  Google Scholar 

  117. Hénon H, Durieu I, Guerouaou D, Lebert F, Pasquier F, Leys D (2001) Poststroke dementia: incidence and relationship to prestroke cognitive decline. Neurology 57:1216–1222. https://doi.org/10.1212/wnl.57.7.1216

    Article  PubMed  Google Scholar 

  118. Hertz L (2008) Bioenergetics of cerebral ischemia: a cellular perspective. Neuropharmacology 55:289–309. https://doi.org/10.1016/j.neuropharm.2008.05.023

    Article  CAS  PubMed  Google Scholar 

  119. Hertz L, Dienel GA (2002) Energy metabolism in the brain. In: Dwyer D (ed) Glucose metabolism in the brain. Academic Press, pp 1–102

    Google Scholar 

  120. Hetz C, Saxena S (2017) ER stress and the unfolded protein response in neurodegeneration. Nat Rev Neurol 13:477–491. https://doi.org/10.1038/nrneurol.2017.99

    Article  CAS  PubMed  Google Scholar 

  121. High FA, Epstein JA (2008) The multifaceted role of Notch in cardiac development and disease. Nat Rev Genet 9:49–61. https://doi.org/10.1038/nrg2279

    Article  CAS  PubMed  Google Scholar 

  122. Hilal S, Chai YL, van Veluw S, Shaik MA, Ikram MK, Venketasubramanian N, Richards AM, Biessels GJ, Chen C (2017) Association between subclinical cardiac biomarkers and clinically manifest cardiac diseases with cortical cerebral microinfarcts. JAMA Neurol 74:403–410

    Article  PubMed  PubMed Central  Google Scholar 

  123. Hilal S, Mok V, Youn YC, Wong A, Ikram MK, Chen CLH (2017) Prevalence, risk factors and consequences of cerebral small vessel diseases: data from three Asian countries. J Neurol Neurosurg Psychiatry 88:669–674. https://doi.org/10.1136/JNNP-2016-315324

    Article  PubMed  Google Scholar 

  124. Ho SC, Woo J, Sham A, Chan SG, Yu ALM (2001) A 3-year follow-up study of social, lifestyle and health predictors of cognitive impairment in a Chinese older cohort. Int J Epidemiol 30:1389–1396. https://doi.org/10.1093/ije/30.6.1389

    Article  CAS  PubMed  Google Scholar 

  125. Hsu M-J, Sheu J-R, Lin C-H, Shen M-Y, Hsu CY (2010) Mitochondrial mechanisms in amyloid beta peptide-induced cerebrovascular degeneration. Biochim Biophys Acta (BBA) Gen Subj 1800:290–296. https://doi.org/10.1016/j.bbagen.2009.08.003

    Article  CAS  Google Scholar 

  126. Hu M, Liu Z, Lv P, Wang H, Zhu Y, Qi Q, Xu J (2017) Autophagy and Akt/CREB signalling play an important role in the neuroprotective effect of nimodipine in a rat model of vascular dementia. Behav Brain Res 325:79–86. https://doi.org/10.1016/j.bbr.2016.11.053

    Article  CAS  PubMed  Google Scholar 

  127. Huang J, Li J, Feng C, Huang X, Wong L, Liu X, Nie Z, Xi G (2018) Blood–brain barrier damage as the starting point of leukoaraiosis caused by cerebral chronic hypoperfusion and its involved mechanisms: effect of agrin and aquaporin-4. Biomed Res Int. https://doi.org/10.1155/2018/2321797

    Article  PubMed  PubMed Central  Google Scholar 

  128. Huang J, Zhang Z, Hong X, Wang J, Wei J, Wen H (2009) Early cognitive predictors of vascular dementia: a population-based longitudinal study in Chinese elderly. J Exp Stroke Transl Med. https://doi.org/10.4172/1939-067x.1000103

    Article  Google Scholar 

  129. Hundal RS, Krssak M, Dufour S, Laurent D, Lebon V, Chandramouli V, Inzucchi SE, Schumann WC, Petersen KF, Landau BR et al (2000) Mechanism by which metformin reduces glucose production in type 2 diabetes. Diabetes 49:2063–2069. https://doi.org/10.2337/diabetes.49.12.2063

    Article  CAS  PubMed  Google Scholar 

  130. Iadecola C (2013) The pathobiology of vascular dementia. Neuron 80:844–866. https://doi.org/10.1016/j.neuron.2013.10.008

    Article  CAS  PubMed  Google Scholar 

  131. Ikram MA, Bersano A, Manso-Calderón R, Jia JP, Schmidt H, Middleton L, Nacmias B, Siddiqi S, Adams HH (2017) Genetics of vascular dementia—review from the ICVD working group. BMC Med 15:48. https://doi.org/10.1186/s12916-017-0813-9

    Article  PubMed  PubMed Central  Google Scholar 

  132. Ingles JL, Wentzel C, Fisk JD, Rockwood K (2002) Neuropsychological predictors of incident dementia in patients with vascular cognitive impairment, without dementia. Stroke 33:1999–2002. https://doi.org/10.1161/01.STR.0000024433.36590.1B

    Article  PubMed  Google Scholar 

  133. Jagtap A, Gawande S, Sharma S (2015) Biomarkers in vascular dementia: a recent update. Biomark Genom Med 7:43–56. https://doi.org/10.1016/j.bgm.2014.11.001

    Article  CAS  Google Scholar 

  134. Jiang XL, Samant S, Lesko LJ, Schmidt S (2015) Clinical pharmacokinetics and pharmacodynamics of clopidogrel. Clin Pharmacokinet 54:147–166. https://doi.org/10.1007/s40262-014-0230-6

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Jørgensen IF, Aguayo-Orozco A, Lademann M, Brunak S (2020) Age-stratified longitudinal study of Alzheimer’s and vascular dementia patients. Alzheimers Dement 16:908–917. https://doi.org/10.1002/alz.12091

    Article  PubMed  PubMed Central  Google Scholar 

  136. Juma W, Lira A, Marzuk A, Marzuk Z, Hakim A, Thompson C (2011) C-reactive protein expression in a rodent model of chronic cerebral hypoperfusion. Brain Res 1414:85–93. https://doi.org/10.1016/j.brainres.2011.07.047

    Article  CAS  PubMed  Google Scholar 

  137. Justin BN, Turek M, Hakim AM (2013) Heart disease as a risk factor for dementia. Clin Epidemiol 5:135–145. https://doi.org/10.2147/clep.S30621

    Article  PubMed  PubMed Central  Google Scholar 

  138. Kakae M, Tobori S, Morishima M, Nagayasu K, Shirakawa H, Kaneko S (2019) Depletion of microglia ameliorates white matter injury and cognitive impairment in a mouse chronic cerebral hypoperfusion model. Biochem Biophys Res Commun 514:1040–1044. https://doi.org/10.1016/j.bbrc.2019.05.055

    Article  CAS  PubMed  Google Scholar 

  139. Kalaria RN (2016) Neuropathological diagnosis of vascular cognitive impairment and vascular dementia with implications for Alzheimer’s disease. Acta Neuropathol 131:659–685. https://doi.org/10.1007/s00401-016-1571-z

    Article  PubMed  PubMed Central  Google Scholar 

  140. Kalaria RN (2018) The pathology and pathophysiology of vascular dementia. Neuropharmacology 134:226–239. https://doi.org/10.1016/j.neuropharm.2017.12.030

    Article  CAS  PubMed  Google Scholar 

  141. Kalogeris T, Baines CP, Krenz M, Korthuis RJ (2012) Cell biology of ischemia/reperfusion injury. Int Rev Cell Mol Biol 298:229–317. https://doi.org/10.1016/B978-0-12-394309-5.00006-7

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Kang JH, Cook N, Manson J, Buring JE, Grodstein F (2007) Low dose aspirin and cognitive function in the women’s health study cognitive cohort. BMJ 334:987. https://doi.org/10.1136/bmj.39166.597836.BE

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Katsura K-I, Kristián T, Smith M-L, Siesjö BK (1994) Acidosis induced by hypercapnia exaggerates ischemic brain damage. J Cereb Blood Flow Metab 14:243–250. https://doi.org/10.1038/jcbfm.1994.31

    Article  CAS  PubMed  Google Scholar 

  144. Kawahara M, Sadakane Y, Koyama H, Konoha K, Ohkawara S (2013) D-histidine and L-histidine attenuate zinc-induced neuronal death in GT1–7 cells. Metallomics Integr Biomet Sci 5(5):453–460

    Article  CAS  Google Scholar 

  145. Kawamura J, Meyer JS, Terayama Y, Weathers S (1991) Leukoaraiosis correlates with cerebral hypoperfusion in vascular dementia. Stroke 22:609–614. https://doi.org/10.1161/01.STR.22.5.609

    Article  CAS  PubMed  Google Scholar 

  146. Kawasaki T, Kawai T (2014) Toll-like receptor signaling pathways. Front Immunol 5:461

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Khan A, Kalaria RN, Corbett A, Ballard C (2016) Update on vascular dementia. J Geriatr Psychiatry Neurol 29:281–301. https://doi.org/10.1177/0891988716654987

    Article  PubMed  Google Scholar 

  148. Kiliaan AJ, Arnoldussen IA, Gustafson DR (2014) Adipokines: a link between obesity and dementia? Lancet Neurol 13:913–923. https://doi.org/10.1016/s1474-4422(14)70085-7

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Killeen MJ, Linder M, Pontoniere P, Crea R (2014) NF-κβ signaling and chronic inflammatory diseases: exploring the potential of natural products to drive new therapeutic opportunities. Drug Discovery Today 19:373–378. https://doi.org/10.1016/j.drudis.2013.11.002

    Article  CAS  PubMed  Google Scholar 

  150. Kim S, Choi SH, Lee YM, Kim MJ, Kim YD, Kim JY, Park JH, Myung W, Na HR, Han HJ et al (2015) Periventricular white matter hyperintensities and the risk of dementia: a CREDOS study. Int Psychogeriatr 27:2069–2077. https://doi.org/10.1017/S1041610215001076

    Article  PubMed  Google Scholar 

  151. Kivipelto M, Mangialasche F, Snyder HM, Allegri R, Andrieu S, Arai H, Baker L, Belleville S, Brodaty H, Brucki SM (2020) World-Wide FINGERS Network: a global approach to risk reduction and prevention of dementia. Alzheimers Dement 16:1078–1094

    Article  PubMed  PubMed Central  Google Scholar 

  152. Kokko JP (1984) Site and mechanism of action of diuretics. Am J Med 77:11–17. https://doi.org/10.1016/s0002-9343(84)80003-0

    Article  CAS  PubMed  Google Scholar 

  153. Kokmen E, Whisnant JP, O’Fallon WM, Chu CP, Beard CM (1996) Dementia after ischemic stroke: a population-based study in Rochester, Minnesota (1960–1984). Neurology 46:154–159. https://doi.org/10.1212/wnl.46.1.154

    Article  CAS  PubMed  Google Scholar 

  154. Kuller LH, Lopez OL, Jagust WJ, Becker JT, DeKosky ST, Lyketsos C, Kawas C, Breitner JC, Fitzpatrick A, Dulberg C (2005) Determinants of vascular dementia in the Cardiovascular Health Cognition Study. Neurology 64:1548–1552. https://doi.org/10.1212/01.Wnl.0000160115.55756.De

    Article  CAS  PubMed  Google Scholar 

  155. Lanza G, Cantone M, Musso S, Borgione E, Scuderi C, Ferri R (2018) Early-onset subcortical ischemic vascular dementia in an adult with mtDNA mutation 3316G>A. J Neurol 265:968–969. https://doi.org/10.1007/s00415-018-8795-x

    Article  PubMed  Google Scholar 

  156. Laurin D, Verreault R, Lindsay J, MacPherson K, Rockwood K (2001) Physical activity and risk of cognitive impairment and dementia in elderly persons. Arch Neurol 58:498–504. https://doi.org/10.1001/archneur.58.3.498

    Article  CAS  PubMed  Google Scholar 

  157. Lautenschlager NT, Cox K, Cyarto EV (2012) The influence of exercise on brain aging and dementia. Biochim Biophys Acta (BBA) Mol Basis Dis 1822:474–481. https://doi.org/10.1016/j.bbadis.2011.07.010

    Article  CAS  Google Scholar 

  158. Leblanc GG, Meschia JF, Stuss DT, Hachinski V (2006) Genetics of vascular cognitive impairment. Stroke 37:248–255. https://doi.org/10.1161/01.STR.0000195177.61184.49

    Article  PubMed  Google Scholar 

  159. Lejri I, Grimm A, Eckert A (2018) Mitochondria, estrogen and female brain aging. Front Aging Neurosci 10:124–124. https://doi.org/10.3389/fnagi.2018.00124

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Lenglet S, Montecucco F, Mach F, Schaller K, Gasche Y, Copin J-C (2017) Analysis of the expression of nine secreted matrix metalloproteinases and their endogenous inhibitors in the brain of mice subjected to ischaemic stroke. Thromb Haemost 112:363–378. https://doi.org/10.1160/TH14-01-0007

    Article  CAS  Google Scholar 

  161. Leuner K, Müller W, Reichert A (2012) From mitochondrial dysfunction to amyloid beta formation: novel insights into the pathogenesis of Alzheimer’s disease. Mol Neurobiol 46:186–193. https://doi.org/10.1007/s12035-012-8307-4

    Article  CAS  PubMed  Google Scholar 

  162. Li H, Liu Y, Lin LT, Wang XR, Du SQ, Yan CQ, He T, Yang JW, Liu CZ (2016) Acupuncture reversed hippocampal mitochondrial dysfunction in vascular dementia rats. Neurochem Int 92:35–42. https://doi.org/10.1016/j.neuint.2015.12.001

    Article  CAS  PubMed  Google Scholar 

  163. Lin J, Wang D, Lan L, Fan Y (2017) Multiple factors involved in the pathogenesis of white matter lesions. Biomed Res Int 2017:9372050. https://doi.org/10.1155/2017/9372050

    Article  PubMed  PubMed Central  Google Scholar 

  164. Lindén T, Skoog I, Fagerberg B, Steen B, Blomstrand C (2004) Cognitive impairment and dementia 20 months after stroke. Neuroepidemiology 23:45–52. https://doi.org/10.1159/000073974

    Article  PubMed  Google Scholar 

  165. Ling C, Liu Z, Song M, Zhang W, Wang S, Liu X, Ma S, Sun S, Fu L, Chu Q et al (2019) Modeling CADASIL vascular pathologies with patient-derived induced pluripotent stem cells. Protein Cell 10:249–271. https://doi.org/10.1007/s13238-019-0608-1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Liu H, Zhang J (2012) Cerebral hypoperfusion and cognitive impairment: the pathogenic role of vascular oxidative stress. Int J Neurosci 122:494–499. https://doi.org/10.3109/00207454.2012.686543

    Article  CAS  PubMed  Google Scholar 

  167. Liu Q, He S, Groysman L, Shaked D, Russin J, Scotton TC, Cen S, Mack WJ (2013) White matter injury due to experimental chronic cerebral hypoperfusion is associated with C5 deposition. PLoS ONE 8:e84802. https://doi.org/10.1371/journal.pone.0084802

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Ma Y, Liu Y, Zhang Z, Yang G-Y (2019) Significance of complement system in ischemic stroke: a comprehensive review. Aging Dis 10:429–462. https://doi.org/10.14336/AD.2019.0119

    Article  PubMed  PubMed Central  Google Scholar 

  169. Magami S, Miyamoto N, Ueno Y, Hira K, Tanaka R, Yamashiro K, Oishi H, Arai H, Urabe T, Hattori N (2019) The effects of astrocyte and oligodendrocyte lineage cell interaction on white matter injury under chronic cerebral hypoperfusion. Neuroscience 406:167–175. https://doi.org/10.1016/j.neuroscience.2019.03.004

    Article  CAS  PubMed  Google Scholar 

  170. Maloney B, Lahiri DK (2016) Epigenetics of dementia: understanding the disease as a transformation rather than a state. Lancet Neurol 15:760–774. https://doi.org/10.1016/S1474-4422(16)00065-X

    Article  CAS  PubMed  Google Scholar 

  171. Man S, Ubogu E, Ransohoff R (2007) Inflammatory cell migration into the central nervous system: a few new twists on an old tale. Brain Pathol 17:243–250. https://doi.org/10.1111/j.1750-3639.2007.00067.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Mancuso C, Scapagini G, Currò D, Stella AMG, Marco CD, Butterfield DA, Calabrese V (2007) Mitochondrial dysfunction, free radical generation and cellular stress response in neurodegenerative disorders. FBL 12:1107–1123

    CAS  Google Scholar 

  173. Markus HS, Schmidt R (2019) Genetics of vascular cognitive impairment. Stroke 50:765–772. https://doi.org/10.1161/STROKEAHA.118.020379

    Article  PubMed  PubMed Central  Google Scholar 

  174. Masumura M, Hata R, Nagai Y, Sawada T (2001) Oligodendroglial cell death with DNA fragmentation in the white matter under chronic cerebral hypoperfusion: comparison between normotensive and spontaneously hypertensive rats. Neurosci Res 39:401–412. https://doi.org/10.1016/S0168-0102(01)00195-X

    Article  CAS  PubMed  Google Scholar 

  175. Mathey E, Park S, Hughes R, Pollard J, Armati P, Barnett M, Taylor B, Dyck P, Kiernan M, Lin C (2015) Chronic inflammatory demyelinating polyradiculoneuropathy: from pathology to phenotype. J Neurol Neurosurg Psychiatry. https://doi.org/10.1136/jnnp-2014-309697

    Article  PubMed  Google Scholar 

  176. Matsushita K, Kuriyama Y, Nagatsuka K, Nakamura M, Sawada T, Omae T (1994) Periventricular white matter lucency and cerebral blood flow autoregulation in hypertensive patients. Hypertension 23:565–568. https://doi.org/10.1161/01.HYP.23.5.565

    Article  CAS  PubMed  Google Scholar 

  177. Matsuyama H, Shindo A, Shimada T, Yata K, Wakita H, Takahashi R, Tomimoto H (2020) Chronic cerebral hypoperfusion activates AIM2 and NLRP3 inflammasome. Brain Res 1736:146779–146779. https://doi.org/10.1016/J.BRAINRES.2020.146779

    Article  CAS  PubMed  Google Scholar 

  178. Matute C (2011) Glutamate and ATP signalling in white matter pathology. J Anat 219:53–64. https://doi.org/10.1111/j.1469-7580.2010.01339.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Matute C, Ransom BR (2012) Roles of white matter in central nervous system pathophysiologies. ASN Neuro 4:e00079–e00079. https://doi.org/10.1042/AN20110060

    Article  PubMed  PubMed Central  Google Scholar 

  180. McAleese KE, Alafuzoff I, Charidimou A, De Reuck J, Grinberg LT, Hainsworth AH, Hortobagyi T, Ince P, Jellinger K, Gao J et al (2016) Post-mortem assessment in vascular dementia: advances and aspirations. BMC Med 14:129–129. https://doi.org/10.1186/s12916-016-0676-5

    Article  PubMed  PubMed Central  Google Scholar 

  181. McQueen J, Reimer M, Holland P, Manso Y, McLaughlin M, Fowler J, Horsburgh K (2014) Restoration of oligodendrocyte pools in a mouse model of chronic cerebral hypoperfusion. PLoS ONE 9:e87227–e87227. https://doi.org/10.1371/journal.pone.0087227

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Mekaj YH, Daci FT, Mekaj AY (2015) New insights into the mechanisms of action of aspirin and its use in the prevention and treatment of arterial and venous thromboembolism. Ther Clin Risk Manag 11:1449–1456. https://doi.org/10.2147/tcrm.S92222

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Miyamoto N, Magami S, Inaba T, Ueno Y, Hira K, Kijima C, Nakajima S, Yamashiro K, Urabe T, Hattori N (2020) The effects of A1/A2 astrocytes on oligodendrocyte linage cells against white matter injury under prolonged cerebral hypoperfusion. Glia 68:1910–1924. https://doi.org/10.1002/glia.23814

    Article  PubMed  Google Scholar 

  184. Miyamoto N, Maki T, Shindo A, Liang AC, Maeda M, Egawa N, Itoh K, Lo EK, Lok J, Ihara M et al (2015) Astrocytes promote oligodendrogenesis after white matter damage via brain-derived neurotrophic factor. J Neurosci 35:14002–14008. https://doi.org/10.1523/jneurosci.1592-15.2015

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Montagne A, Nikolakopoulou AM, Zhao Z, Sagare AP, Si G, Lazic D, Barnes SR, Daianu M, Ramanathan A, Go A et al (2018) Pericyte degeneration causes white matter dysfunction in the mouse central nervous system. Nat Med 24(3):326–337. https://doi.org/10.1038/nm.4482

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Morris MC (2016) Nutrition and risk of dementia: overview and methodological issues. Ann N Y Acad Sci 1367:31–37. https://doi.org/10.1111/nyas.13047

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Morris MC (2012) Nutritional determinants of cognitive aging and dementia. Proc Nutr Soc 71:1–13. https://doi.org/10.1017/s0029665111003296

    Article  CAS  PubMed  Google Scholar 

  188. Morris MC, Tangney CC (2014) Dietary fat composition and dementia risk. Neurobiol Aging 35(Suppl 2):S59-64. https://doi.org/10.1016/j.neurobiolaging.2014.03.038

    Article  CAS  PubMed  Google Scholar 

  189. Mracskó E, Hugyecz M, Institoris A, Farkas E, Bari F (2009) Changes in pro-oxidant and antioxidant enzyme levels during cerebral hypoperfusion in rats. Brain Res 1321:13–19. https://doi.org/10.1016/j.brainres.2009.11.080

    Article  CAS  PubMed  Google Scholar 

  190. Nagai M, Hoshide S, Kario K (2010) Hypertension and dementia. Am J Hypertens 23:116–124. https://doi.org/10.1038/ajh.2009.212

    Article  PubMed  Google Scholar 

  191. Nation DA, Sweeney MD, Montagne A, Sagare AP, D’Orazio LM, Pachicano M, Sepehrband F, Nelson AR, Buennagel DP, Harrington MG et al (2019) Blood–brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nat Med 25(2):270–276. https://doi.org/10.1038/s41591-018-0297-y

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Navarro A, Boveris A (2007) The mitochondrial energy transduction system and the aging process. Am J Physiol Cell Physiol 292:C670–C686. https://doi.org/10.1152/ajpcell.00213.2006

    Article  CAS  PubMed  Google Scholar 

  193. Newman AB, Fitzpatrick AL, Lopez O, Jackson S, Lyketsos C, Jagust W, Ives D, Dekosky ST, Kuller LH (2005) Dementia and Alzheimer’s disease incidence in relationship to cardiovascular disease in the Cardiovascular Health Study cohort. J Am Geriatr Soc 53:1101–1107. https://doi.org/10.1111/j.1532-5415.2005.53360.x

    Article  PubMed  Google Scholar 

  194. Nishio K, Ihara M, Yamasaki N, Kalaria R, Maki T, Fujita Y, Ito H, Oishi N, Fukuyama H, Miyakawa T et al (2010) A mouse model characterizing features of vascular dementia with hippocampal atrophy. Stroke J Cereb Circ 41:1278–1284. https://doi.org/10.1161/STROKEAHA.110.581686

    Article  Google Scholar 

  195. Niu XL, Jiang X, Xu GD, Zheng GM, Tang ZP, Yin N, Li XQ, Yang YY, Lv PY (2018) DL-3-n-butylphthalide alleviates vascular cognitive impairment by regulating endoplasmic reticulum stress and the Shh/Ptch1 signaling-pathway in rats. J Cell Physiol. https://doi.org/10.1002/jcp.27332

    Article  PubMed  Google Scholar 

  196. Nunnari J, Suomalainen A (2012) Mitochondria: in sickness and in health. Cell 148:1145–1159. https://doi.org/10.1016/j.cell.2012.02.035

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. O’Barr S, Cooper NR (2000) The C5a complement activation peptide increases IL-1β and IL-6 release from amyloid-β primed human monocytes: implications for Alzheimer’s disease. J Neuroimmunol 109:87–94. https://doi.org/10.1016/S0165-5728(00)00291-5

    Article  PubMed  Google Scholar 

  198. O’Sullivan M, Lythgoe DJ, Pereira AC, Summers PE, Jarosz JM, Williams SCR, Markus HS (2002) Patterns of cerebral blood flow reduction in patients with ischemic leukoaraiosis. Neurology 59:321–326. https://doi.org/10.1212/WNL.59.3.321

    Article  PubMed  Google Scholar 

  199. Ogino S, Lochhead P, Chan AT, Nishihara R, Cho E, Wolpin BM, Meyerhardt JA, Meissner A, Schernhammer ES, Fuchs CS et al (2013) Molecular pathological epidemiology of epigenetics: emerging integrative science to analyze environment, host, and disease. Mod Pathol Off J US Can Acad Pathol Inc 26:465–484. https://doi.org/10.1038/modpathol.2012.214

    Article  CAS  Google Scholar 

  200. Ott A, Breteler MM, van Harskamp F, Claus JJ, van der Cammen TJ, Grobbee DE, Hofman A (1995) Prevalence of Alzheimer’s disease and vascular dementia: association with education. The Rotterdam study. BMJ 310:970–973. https://doi.org/10.1136/bmj.310.6985.970

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Ozawa M, Ninomiya T, Ohara T, Doi Y, Uchida K, Shirota T, Yonemoto K, Kitazono T, Kiyohara Y (2013) Dietary patterns and risk of dementia in an elderly Japanese population: the Hisayama Study. Am J Clin Nutr 97:1076–1082. https://doi.org/10.3945/ajcn.112.045575

    Article  CAS  PubMed  Google Scholar 

  202. Paciaroni M, Bogousslavsky J (2013) Connecting cardiovascular disease and dementia: further evidence. J Am Heart Assoc 2:e000656. https://doi.org/10.1161/jaha.113.000656

    Article  PubMed  PubMed Central  Google Scholar 

  203. Park J-M, Kim YJ, Song MK, Lee J-M, Kim Y-J (2018) Genome-wide DNA methylation profiling in a rat model with vascular dementia. Mol Med Rep 18:123–130. https://doi.org/10.3892/mmr.2018.8990

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Peila R, Rodriguez BL, Launer LJ (2002) Type 2 diabetes, APOE gene, and the risk for dementia and related pathologies: the Honolulu-Asia aging study. Diabetes 51:1256–1262. https://doi.org/10.2337/diabetes.51.4.1256

    Article  CAS  PubMed  Google Scholar 

  205. Phillips NA, Mate-Kole CC (1997) Cognitive deficits in peripheral vascular disease. A comparison of mild stroke patients and normal control subjects. Stroke 28:777–784. https://doi.org/10.1161/01.str.28.4.777

    Article  CAS  PubMed  Google Scholar 

  206. Poh L, Fann DY, Wong P, Lim HM, Foo SL, Kang S-W, Rajeev V, Selvaraji S, Iyer VR, Parathy N et al (2021) AIM2 inflammasome mediates hallmark neuropathological alterations and cognitive impairment in a mouse model of vascular dementia. Mol Psychiatry 26:4544–4560. https://doi.org/10.1038/s41380-020-00971-5

    Article  CAS  PubMed  Google Scholar 

  207. Poh L, Razak S, Lim HM, Lai MKP, Chen CL, Lim LHK, Arumugam TV, Fann DY (2021) AIM2 inflammasome mediates apoptotic and pyroptotic death in the cerebellum following chronic hypoperfusion. Exp Neurol 346:113856. https://doi.org/10.1016/j.expneurol.2021.113856

    Article  CAS  PubMed  Google Scholar 

  208. Portegies ML, Wolters FJ, Hofman A, Ikram MK, Koudstaal PJ, Ikram MA (2016) Prestroke vascular pathology and the risk of recurrent stroke and poststroke dementia. Stroke 47:2119–2122. https://doi.org/10.1161/strokeaha.116.014094

    Article  PubMed  Google Scholar 

  209. Posner HB, Tang MX, Luchsinger J, Lantigua R, Stern Y, Mayeux R (2002) The relationship of hypertension in the elderly to AD, vascular dementia, and cognitive function. Neurology 58:1175–1181. https://doi.org/10.1212/wnl.58.8.1175

    Article  CAS  PubMed  Google Scholar 

  210. Praticò D, Clark CM, Liun F, Lee VYM, Trojanowski JQ (2002) Increase of brain oxidative stress in mild cognitive impairment: a possible predictor of Alzheimer disease. Arch Neurol 59:972–976. https://doi.org/10.1001/archneur.59.6.972

    Article  PubMed  Google Scholar 

  211. Price BR, Wilcock DM, Weekman EM (2018) Hyperhomocysteinemia as a risk factor for vascular contributions to cognitive impairment and dementia. Front Aging Neurosci 10:350–350. https://doi.org/10.3389/fnagi.2018.00350

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Rafnsson SB, Deary IJ, Fowkes FG (2009) Peripheral arterial disease and cognitive function. Vasc Med 14:51–61. https://doi.org/10.1177/1358863x08095027

    Article  PubMed  Google Scholar 

  213. Rajeev V, Fann DY, Dinh QN, Kim HA, De Silva TM, Jo DG, Drummond GR, Sobey CG, Lai MKP, Chen CL et al (2022) Intermittent fasting attenuates Hallmark vascular and neuronal pathologies in a mouse model of vascular cognitive impairment. Int J Biol Sci 18:6052–6067. https://doi.org/10.7150/ijbs.75188

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Rajeev V, Fann DY, Dinh QN, Kim HA, De Silva TM, Lai MKP, Chen CL, Drummond GR, Sobey CG, Arumugam TV (2022) Pathophysiology of blood brain barrier dysfunction during chronic cerebral hypoperfusion in vascular cognitive impairment. Theranostics 12:1639–1658. https://doi.org/10.7150/thno.68304

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Ravaglia G, Forti P, Maioli F, Chiappelli M, Montesi F, Tumini E, Mariani E, Licastro F, Patterson C (2007) Blood inflammatory markers and risk of dementia: the conselice study of brain aging. Neurobiol Aging 28:1810–1820. https://doi.org/10.1016/j.neurobiolaging.2006.08.012

    Article  CAS  PubMed  Google Scholar 

  216. Ravona-Springer R, Schnaider-Beeri M (2011) The association of diabetes and dementia and possible implications for nondiabetic populations. Expert Rev Neurother 11:1609–1617. https://doi.org/10.1586/ern.11.152

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Reimer M, McQueen J, Searcy L, Scullion G, Desmazières A, Holland P, Smith J, Gliddon C, Wood E, Herzyk P et al (2011) Rapid disruption of axon-glial integrity in response to mild cerebral hypoperfusion. J Neurosci Off J Soc Neurosci 31:18185–18194. https://doi.org/10.1523/JNEUROSCI.4936-11.2011

    Article  CAS  Google Scholar 

  218. Rizzuto R, De Stefani D, Raffaello A, Mammucari C (2012) Mitochondria as sensors and regulators of calcium signalling. Nat Rev Mol Cell Biol 13:566–578. https://doi.org/10.1038/nrm3412

    Article  CAS  PubMed  Google Scholar 

  219. Roberts RO, Knopman DS, Geda YE, Cha RH, Roger VL, Petersen RC (2010) Coronary heart disease is associated with non-amnestic mild cognitive impairment. Neurobiol Aging 31:1894–1902. https://doi.org/10.1016/j.neurobiolaging.2008.10.018

    Article  PubMed  Google Scholar 

  220. Rohn TT, McCarty KL, Love JE, Head E (2014) Is apolipoprotein E4 an important risk factor for dementia in persons with down syndrome? J Parkinson’s Dis Alzheimer’s Dis 1:7–7. https://doi.org/10.13188/2376-922x.1000004

    Article  Google Scholar 

  221. Román GC (2003) Stroke, cognitive decline and vascular dementia: the silent epidemic of the 21st century. Neuroepidemiology 22:161–164. https://doi.org/10.1159/000069885

    Article  PubMed  Google Scholar 

  222. Román GC (2002) Vascular dementia may be the most common form of dementia in the elderly. J Neurol Sci 203:7–10. https://doi.org/10.1016/S0022-510X(02)00252-6

    Article  PubMed  Google Scholar 

  223. Rosenberg GA, Sullivan N, Esiri MM (2001) White matter damage is associated with matrix metalloproteinases in vascular dementia. Stroke 32:1162–1167. https://doi.org/10.1161/01.STR.32.5.1162

    Article  CAS  PubMed  Google Scholar 

  224. Ruitenberg A, Den Heijer T, Bakker SLM, Van Swieten JC, Koudstaal PJ, Hofman A, Breteler MMB (2005) Cerebral hypoperfusion and clinical onset of dementia: the Rotterdam study. Ann Neurol 57:789–794. https://doi.org/10.1002/ANA.20493

    Article  PubMed  Google Scholar 

  225. Russ TC, Stamatakis E, Hamer M, Starr JM, Kivimäki M, Batty GD (2013) Socioeconomic status as a risk factor for dementia death: individual participant meta-analysis of 86508 men and women from the UK. Br J Psychiatry 203:10–17. https://doi.org/10.1192/bjp.bp.112.119479

    Article  PubMed  PubMed Central  Google Scholar 

  226. Rygiel K (2016) Can angiotensin-converting enzyme inhibitors impact cognitive decline in early stages of Alzheimer’s disease? An overview of research evidence in the elderly patient population. J Postgrad Med 62:242–248. https://doi.org/10.4103/0022-3859.188553

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  227. Sabayan B, Jansen S, Oleksik AM, van Osch MJP, van Buchem MA, van Vliet P, de Craen AJM, Westendorp RGJ (2012) Cerebrovascular hemodynamics in Alzheimer’s disease and vascular dementia: a meta-analysis of transcranial Doppler studies. Ageing Res Rev 11:271–277. https://doi.org/10.1016/j.arr.2011.12.009

    Article  PubMed  Google Scholar 

  228. Sachdev P, Kalaria R, O’Brien J, Skoog I, Alladi S, Black SE, Blacker D, Blazer D, Chen C, Chui H (2014) Diagnostic criteria for vascular cognitive disorders: a VASCOG statement. Alzheimer Dis Assoc Disord 28:206–206

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Saito A (2014) Physiological functions of endoplasmic reticulum stress transducer OASIS in central nervous system. Anat Sci Int 89:11–20. https://doi.org/10.1007/s12565-013-0214-x

    Article  CAS  PubMed  Google Scholar 

  230. Saridin FN, Hilal S, Villaraza SG, Reilhac A, Gyanwali B, Tanaka T, Stephenson MC, Ng SL, Vrooman H, van der Flier WM et al (2020) Brain amyloid β, cerebral small vessel disease, and cognition. Neurology 95:e2845–e2853. https://doi.org/10.1212/WNL.0000000000011029

    Article  CAS  PubMed  Google Scholar 

  231. Scheel P, Puls I, Becker G, Schöning M (1999) Volume reduction in cerebral blood flow in patients with vascular dementia. Lancet 354:2137–2137. https://doi.org/10.1016/S0140-6736(99)04016-7

    Article  CAS  PubMed  Google Scholar 

  232. Schmidt R, Schmidt H, Curb J, Masaki K, White L, Launer L (2002) Early inflammation and dementia: a 25-year follow-up of the Honolulu-Asia aging study. Ann Neurol 52:168–174. https://doi.org/10.1002/ana.10265

    Article  PubMed  Google Scholar 

  233. Schmitz T, Chew L-J (2008) Cytokines and myelination in the central nervous system. TheScientificWorldJOURNAL 8:1119–1147. https://doi.org/10.1100/tsw.2008.140

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Schuff N, Matsumoto S, Kmiecik J, Studholme C, Du A, Ezekiel F, Miller BL, Kramer JH, Jagust WJ, Chui HC et al (2009) Cerebral blood flow in ischemic vascular dementia and Alzheimer’s disease, measured by arterial spin-labeling magnetic resonance imaging. Alzheimers Dement 5:454–462. https://doi.org/10.1016/J.JALZ.2009.04.1233

    Article  PubMed  PubMed Central  Google Scholar 

  235. Schuit AJ, Feskens MEJ, Launer LJ, Kromhout D (2001) Physical activity and cognitive decline, the role of the apolipoprotein e4 allele. Med Sci Sports Exerc 33:772

    Article  CAS  PubMed  Google Scholar 

  236. Schwarzinger M, Pollock BG, Hasan OSM, Dufouil C, Rehm J (2018) Contribution of alcohol use disorders to the burden of dementia in France 2008–13: a nationwide retrospective cohort study. Lancet Public Health 3:e124–e132. https://doi.org/10.1016/s2468-2667(18)30022-7

    Article  PubMed  Google Scholar 

  237. Selvaraji S, Efthymios M, Foo RSY, Fann DY, Lai MKP, Chen CLH, Lim KL, Arumugam TV (2022) Time-restricted feeding modulates the DNA methylation landscape, attenuates hallmark neuropathology and cognitive impairment in a mouse model of vascular dementia. Theranostics 12:3007–3023. https://doi.org/10.7150/thno.71815

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. Shabir O, Berwick J, Francis S (2018) Neurovascular dysfunction in vascular dementia, Alzhiemers and atherosclerosis. BMC Neurosci. https://doi.org/10.1186/s12868-018-0465-5

    Article  PubMed  PubMed Central  Google Scholar 

  239. Sharp ES, Gatz M (2011) Relationship between education and dementia: an updated systematic review. Alzheimer Dis Assoc Disord 25:289–304. https://doi.org/10.1097/WAD.0b013e318211c83c

    Article  PubMed  PubMed Central  Google Scholar 

  240. Shen Y, Halperin JA, Lee C-M (1995) Complement-mediated neurotoxicity is regulated by homologous restriction. Brain Res 671:282–292. https://doi.org/10.1016/0006-8993(94)01264-I

    Article  CAS  PubMed  Google Scholar 

  241. Shi G-X, Liu C-Z, Wang L-P, Guan L-P, Li S-Q (2012) Biomarkers of oxidative stress in vascular dementia patients. Can J Neurol Sci J Can Sci Neurol 39:65–68. https://doi.org/10.1017/S0317167100012701

    Article  Google Scholar 

  242. Shibata M, Ohtani R, Ihara M, Tomimoto H (2004) White matter lesions and glial activation in a novel mouse model of chronic cerebral hypoperfusion. Stroke 35:2598–2603. https://doi.org/10.1161/01.Str.0000143725.19053.60

    Article  PubMed  Google Scholar 

  243. Shibata M, Yamasaki N, Miyakawa T, Kalaria RN, Fujita Y, Ohtani R, Ihara M, Takahashi R, Tomimoto H (2007) Selective impairment of working memory in a mouse model of chronic cerebral hypoperfusion. Stroke 38:2826–2832. https://doi.org/10.1161/STROKEAHA.107.490151

    Article  PubMed  Google Scholar 

  244. Silva IVG, de Figueiredo RC, Rios DRA (2019) Effect of different classes of antihypertensive drugs on endothelial function and inflammation. Int J Mol Sci. https://doi.org/10.3390/ijms20143458

    Article  PubMed  PubMed Central  Google Scholar 

  245. Simpson JE, Fernando MS, Clark L, Ince PG, Matthews F, Forster G, O’Brien JT, Barber R, Kalaria RN, Brayne C et al (2007) White matter lesions in an unselected cohort of the elderly: astrocytic, microglial and oligodendrocyte precursor cell responses. Neuropathol Appl Neurobiol 33:410–419. https://doi.org/10.1111/j.1365-2990.2007.00828.x

    Article  CAS  PubMed  Google Scholar 

  246. Skoog I (1998) Status of risk factors for vascular dementia. Neuroepidemiology 17:2–9. https://doi.org/10.1159/000026147

    Article  CAS  PubMed  Google Scholar 

  247. Skrobot OA, Attems J, Esiri M, Hortobágyi T, Ironside JW, Kalaria RN, King A, Lammie GA, Mann D, Neal J et al (2016) Vascular cognitive impairment neuropathology guidelines (VCING): the contribution of cerebrovascular pathology to cognitive impairment. Brain 139:2957–2969. https://doi.org/10.1093/brain/aww214

    Article  PubMed  Google Scholar 

  248. Sofroniew MV (2014) Astrogliosis. Cold Spring Harb Perspect Biol 7:a020420–a020420. https://doi.org/10.1101/cshperspect.a020420

    Article  PubMed  Google Scholar 

  249. Son H-H, Pt OhMS, Jung L, Pt PMS, Rae J, Pt PhD (2010) The effect of an exercise program on activities of daily living (ADL), balance and cognition in elderly individuals with Alzheimer’s disease and vascular dementia. J Korean Phys Ther 22:53–60

    Google Scholar 

  250. Stamatovic SM, Johnson AM, Keep RF, Andjelkovic AV (2016) Junctional proteins of the blood–brain barrier: new insights into function and dysfunction. Tissue Barriers. https://doi.org/10.1080/21688370.2016.1154641

    Article  PubMed  PubMed Central  Google Scholar 

  251. Stampfer MJ, Willett WC, Colditz GA, Rosner B, Speizer FE, Hennekens CH (1985) A prospective study of postmenopausal estrogen therapy and coronary heart disease. N Engl J Med 313:1044–1049. https://doi.org/10.1056/nejm198510243131703

    Article  CAS  PubMed  Google Scholar 

  252. Stanimirovic DB, Friedman A (2012) Pathophysiology of the neurovascular unit: disease cause or consequence? J Cereb Blood Flow Metab Off J Int Soc Cereb Blood Flow Metabol 32:1207–1221. https://doi.org/10.1038/jcbfm.2012.25

    Article  CAS  Google Scholar 

  253. Stern Y, Gurland B, Tatemichi TK, Tang MX, Wilder D, Mayeux R (1994) Influence of education and occupation on the incidence of Alzheimer’s disease. JAMA 271:1004–1010

    Article  CAS  PubMed  Google Scholar 

  254. Stewart R (2002) Vascular dementia: a diagnosis running out of time. Br J Psychiatry 180:152–156. https://doi.org/10.1192/bjp.180.2.152

    Article  PubMed  Google Scholar 

  255. Stys PK, Waxman SG, Ransom BR (1992) Ionic mechanisms of anoxic injury in mammalian CNS white matter: role of Na+ channels and Na(+)–Ca2+ exchanger. J Neurosci 12:430–439. https://doi.org/10.1523/JNEUROSCI.12-02-00430.1992

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  256. Sudduth TL, Powell DK, Smith CD, Greenstein A, Wilcock DM (2013) Induction of hyperhomocysteinemia models vascular dementia by induction of cerebral microhemorrhages and neuroinflammation. J Cereb Blood Flow Metab 33:708–715. https://doi.org/10.1038/jcbfm.2013.1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  257. Sun Z-K, Ma XR, Jia YJ, Liu YR, Zhang JW, Zhang BA (2014) Effects of resveratrol on apoptosis in a rat model of vascular dementia. Exp Ther Med 7:843–848. https://doi.org/10.3892/etm.2014.1542

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  258. Sweeney MD, Montagne A, Sagare AP, Nation DA, Schneider LS, Chui HC, Harrington MG, Pa J, Law M, Wang DJJ (2019) Vascular dysfunction—the disregarded partner of Alzheimer’s disease. Alzheimers Dement 15:158–167

    Article  PubMed  PubMed Central  Google Scholar 

  259. Takeshita Y, Ransohoff RM (2012) Inflammatory cell trafficking across the blood-brain barrier: chemokine regulation and in vitro models. Immunol Rev 248:228–239. https://doi.org/10.1111/j.1600-065X.2012.01127.x

    Article  PubMed  PubMed Central  Google Scholar 

  260. Takeuchi O, Akira S (2010) Pattern recognition receptors and inflammation. Cell 140:805–820. https://doi.org/10.1016/j.cell.2010.01.022

    Article  CAS  PubMed  Google Scholar 

  261. Tampi RR, Tampi DJ, Balachandran S, Srinivasan S (2016) Antipsychotic use in dementia: a systematic review of benefits and risks from meta-analyses. Ther Adv Chronic Dis 7:229–245. https://doi.org/10.1177/2040622316658463

    Article  PubMed  PubMed Central  Google Scholar 

  262. Tan R, Traylor M, Rutten-Jacobs L, Markus H (2017) New insights into mechanisms of small vessel disease stroke from genetics. Clin Sci 131:515–531. https://doi.org/10.1042/cs20160825

    Article  CAS  Google Scholar 

  263. Tanaka K-I, Kawahara M (2017) Copper enhances zinc-induced neurotoxicity and the endoplasmic reticulum stress response in a neuronal model of vascular dementia. Front Neurosci 11:58–58. https://doi.org/10.3389/fnins.2017.00058

    Article  PubMed  PubMed Central  Google Scholar 

  264. Tang D, Kang R, Berghe TV, Vandenabeele P, Kroemer G (2019) The molecular machinery of regulated cell death. Cell Res 29:347–364. https://doi.org/10.1038/s41422-019-0164-5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  265. Tanovic A, Alfaro V (2006) Glutamate-related excitotoxicity neuroprotection with memantine, an uncompetitive antagonist of NMDA-glutamate receptor, in Alzheimer’s disease and vascular dementia. Rev Neurol 42:607–616. https://doi.org/10.33588/rn.4210.2005252

    Article  CAS  PubMed  Google Scholar 

  266. Tarumi T, Shah F, Tanaka H, Haley AP (2011) Association between central elastic artery stiffness and cerebral perfusion in deep subcortical gray and white matter. Am J Hypertens 24:1108–1113. https://doi.org/10.1038/ajh.2011.101

    Article  CAS  PubMed  Google Scholar 

  267. Tasci I, Safer U, Naharci MI, Gezer M, Demir O, Bozoglu E, Doruk H (2018) Undetected peripheral arterial disease among older adults with Alzheimer’s disease and other dementias. Am J Alzheimers Dis Other Demen 33:5–11. https://doi.org/10.1177/1533317517724000

    Article  PubMed  Google Scholar 

  268. Tham W, Auchus AP, Thong M, Goh M-L, Chang H-M, Wong M-C, Chen CPLH (2002) Progression of cognitive impairment after stroke: one year results from a longitudinal study of Singaporean stroke patients. J Neurol Sci 203:49–52. https://doi.org/10.1016/S0022-510X(02)00260-5

    Article  PubMed  Google Scholar 

  269. Tomimoto H, Akiguchi I, Suenaga T, Nishimura M, Wakita H, Nakamura S, Kimura J (1996) Alterations of the blood–brain barrier and glial cells in white-matter lesions in cerebrovascular and Alzheimer’s disease patients. Stroke 27:2069–2074. https://doi.org/10.1161/01.STR.27.11.2069

    Article  CAS  PubMed  Google Scholar 

  270. Topiwala A, Allan CL, Valkanova V, Zsoldos E, Filippini N, Sexton C, Mahmood A, Fooks P, Singh-Manoux A, Mackay CE et al (2017) Moderate alcohol consumption as risk factor for adverse brain outcomes and cognitive decline: longitudinal cohort study. BMJ 357:j2353. https://doi.org/10.1136/bmj.j2353

    Article  PubMed  PubMed Central  Google Scholar 

  271. Toth P, Tarantini S, Csiszar A, Ungvari Z (2017) Functional vascular contributions to cognitive impairment and dementia: mechanisms and consequences of cerebral autoregulatory dysfunction, endothelial impairment, and neurovascular uncoupling in aging. Am J Physiol Heart Circ Physiol 312:H1–H20. https://doi.org/10.1152/AJPHEART.00581.2016

    Article  PubMed  Google Scholar 

  272. Toyama K, Koibuchi N, Uekawa K, Hasegawa Y, Kataoka K, Katayama T, Sueta D, Ma MJ, Nakagawa T, Yasuda O et al (2014) Apoptosis signal-regulating kinase 1 is a novel target molecule for cognitive impairment induced by chronic cerebral hypoperfusion. Arterioscler Thromb Vasc Biol 34:616–625. https://doi.org/10.1161/ATVBAHA.113.302440

    Article  CAS  PubMed  Google Scholar 

  273. Tripolt NJ, Stekovic S, Aberer F, Url J, Pferschy PN, Schröder S, Verheyen N, Schmidt A, Kolesnik E, Narath SH et al (2018) Intermittent fasting (alternate day fasting) in healthy, non-obese adults: protocol for a cohort trial with an embedded randomized controlled pilot trial. Adv Ther 35:1265–1283. https://doi.org/10.1007/s12325-018-0746-5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  274. Troen AM, Shea-Budgell M, Shukitt-Hale B, Smith DE, Selhub J, Rosenberg IH (2008) B-vitamin deficiency causes hyperhomocysteinemia and vascular cognitive impairment in mice. Proc Natl Acad Sci U S A 105:12474–12479. https://doi.org/10.1073/pnas.0805350105

    Article  PubMed  PubMed Central  Google Scholar 

  275. Troncoso JC, Zonderman AB, Resnick SM, Crain B, Pletnikova O, O’Brien RJ (2008) Effect of infarcts on dementia in the Baltimore longitudinal study of aging. Ann Neurol 64:168–176. https://doi.org/10.1002/ana.21413

    Article  PubMed  PubMed Central  Google Scholar 

  276. Truettner JS, Alonso OF, Dietrich WD (2005) Influence of therapeutic hypothermia on matrix metalloproteinase activity after traumatic brain injury in rats. J Cereb Blood Flow Metab 25:1505–1516. https://doi.org/10.1038/SJ.JCBFM.9600150

    Article  CAS  PubMed  Google Scholar 

  277. Ueno M, Tomimoto H, Akiguchi I, Wakita H, Sakamoto H (2016) Blood–brain barrier disruption in white matter lesions in a rat model of chronic cerebral hypoperfusion. J Cereb Blood Flow Metab 22:97–104. https://doi.org/10.1097/00004647-200201000-00012

    Article  Google Scholar 

  278. Ungvari Z, Kaley G, de Cabo R, Sonntag WE, Csiszar A (2010) Mechanisms of vascular aging: new perspectives. J Gerontol A Biol Sci Med Sci 65:1028–1041. https://doi.org/10.1093/gerona/glq113

    Article  PubMed  Google Scholar 

  279. Utech M, Mennigen R, Bruewer M (2010) Endocytosis and recycling of tight junction proteins in inflammation. J Biomed Biotechnol. https://doi.org/10.1155/2010/484987

    Article  PubMed  Google Scholar 

  280. van de Rest O, Berendsen AA, Haveman-Nies A, de Groot LC (2015) Dietary patterns, cognitive decline, and dementia: a systematic review. Adv Nutr 6:154–168. https://doi.org/10.3945/an.114.007617

    Article  PubMed  PubMed Central  Google Scholar 

  281. van der Flier WM, Skoog I, Schneider JA, Pantoni L, Mok V, Chen CLH, Scheltens P (2018) Vascular cognitive impairment. Nat Rev Dis Primers 4(1):1–16. https://doi.org/10.1038/nrdp.2018.3

    Article  Google Scholar 

  282. Varatharaj A, Galea I (2017) The blood–brain barrier in systemic inflammation. Brain Behav Immun 60:1–12. https://doi.org/10.1016/j.bbi.2016.03.010

    Article  CAS  PubMed  Google Scholar 

  283. Venkat P, Chopp M, Chen J (2015) Models and mechanisms of vascular dementia. Exp Neurol 272:97–108. https://doi.org/10.1016/J.EXPNEUROL.2015.05.006

    Article  PubMed  PubMed Central  Google Scholar 

  284. Verbaten MN (2009) Chronic effects of low to moderate alcohol consumption on structural and functional properties of the brain: beneficial or not? Hum Psychopharmacol 24:199–205. https://doi.org/10.1002/hup.1022

    Article  CAS  PubMed  Google Scholar 

  285. Verdelho A, Madureira S, Ferro JM, Baezner H, Blahak C, Poggesi A, Hennerici M, Pantoni L, Fazekas F, Scheltens P et al (2012) Physical activity prevents progression for cognitive impairment and vascular dementia. Stroke 43:3331–3335. https://doi.org/10.1161/STROKEAHA.112.661793

    Article  PubMed  Google Scholar 

  286. Vernooij MW, Van der Lugt A, Ikram MA, Wielopolski PA, Vrooman HA, Hofman A, Krestin GP, Breteler MM (2007) Total cerebral blood flow and total brain perfusion in the general population: the Rotterdam scan study. J Cereb Blood Flow Metab 28:412–419. https://doi.org/10.1038/SJ.JCBFM.9600526

    Article  PubMed  Google Scholar 

  287. Wallace DC (2001) A mitochondrial paradigm for degenerative diseases and ageing. Novartis Found Symp 235:247–263. https://doi.org/10.1002/0470868694.ch20

    Article  CAS  PubMed  Google Scholar 

  288. Wallin A, Kapaki E, Boban M, Engelborghs S, Hermann DM, Huisa B, Jonsson M, Kramberger MG, Lossi L, Malojcic B et al (2017) Biochemical markers in vascular cognitive impairment associated with subcortical small vessel disease—a consensus report. BMC Neurol 17:102–102. https://doi.org/10.1186/s12883-017-0877-3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  289. Wang S, Cao C, Chen Z, Bankaitis V, Tzima E, Sheibani N, Burridge K (2012) Pericytes regulate vascular basement membrane remodeling and govern neutrophil extravasation during inflammation. PLoS ONE 7:e45499. https://doi.org/10.1371/journal.pone.0045499

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  290. Wang T, Liu CZ, Yu JC, Jiang W, Han JX (2009) Acupuncture protected cerebral multi-infarction rats from memory impairment by regulating the expression of apoptosis related genes Bcl-2 and Bax in hippocampus. Physiol Behav 96:155–161. https://doi.org/10.1016/j.physbeh.2008.09.024

    Article  CAS  PubMed  Google Scholar 

  291. Ward NC, Watts GF, Eckel RH (2019) Statin toxicity. Circ Res 124:328–350. https://doi.org/10.1161/circresaha.118.312782

    Article  CAS  PubMed  Google Scholar 

  292. Wardlaw JM, Doubal F, Armitage P, Chappell F, Carpenter T, Maniega SM, Farrall A, Sudlow C, Dennis M, Dhillon B (2009) Lacunar stroke is associated with diffuse blood–brain barrier dysfunction. Ann Neurol 65:194–202. https://doi.org/10.1002/ANA.21549

    Article  PubMed  Google Scholar 

  293. Webb AJS, Simoni M, Mazzucco S, Kuker W, Schulz U, Rothwell PM (2012) Increased cerebral arterial pulsatility in patients with leukoaraiosis. Stroke 43:2631–2636. https://doi.org/10.1161/STROKEAHA.112.655837

    Article  PubMed  Google Scholar 

  294. Wentzel C, Rockwood K, MacKnight C, Hachinski V, Hogan DB, Feldman H, Østbye T, Wolfson C, Gauthier S, Verreault R et al (2001) Progression of impairment in patients with vascular cognitive impairment without dementia. Neurology 57:714–716. https://doi.org/10.1212/WNL.57.4.714

    Article  CAS  PubMed  Google Scholar 

  295. Wiysonge CS, Bradley HA, Volmink J, Mayosi BM, Opie LH (2017) Beta-blockers for hypertension. Cochrane Database Syst Rev 1:Cd002003. https://doi.org/10.1002/14651858.CD002003.pub5

    Article  PubMed  Google Scholar 

  296. Wu C-x, Liu R, Gao M, Zhao G, Wu S, Wu C-f, Du G-h (2013) Pinocembrin protects brain against ischemia/reperfusion injury by attenuating endoplasmic reticulum stress induced apoptosis. Neurosci Lett 546:57–62. https://doi.org/10.1016/j.neulet.2013.04.060

    Article  CAS  PubMed  Google Scholar 

  297. Wu L-Y, Kan CN, Cheah IK, Chong JR, Xu X, Vrooman H, Hilal S, Venketasubramanian N, Chen CP, Halliwell B et al (2022) Low plasma ergothioneine predicts cognitive and functional decline in an elderly cohort attending memory clinics. Antioxidants 11:1717

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  298. Wu LY, Cheah IK, Chong JR, Chai YL, Tan JY, Hilal S, Vrooman H, Chen CP, Halliwell B, Lai MKP (2021) Low plasma ergothioneine levels are associated with neurodegeneration and cerebrovascular disease in dementia. Free Radic Biol Med 177:201–211. https://doi.org/10.1016/j.freeradbiomed.2021.10.019

    Article  CAS  PubMed  Google Scholar 

  299. Wu X, Sun J, Li L (2013) Chronic cerebrovascular hypoperfusion affects global DNA methylation and histone acetylation in rat brain. Neurosci Bull 29:685–692. https://doi.org/10.1007/s12264-013-1345-8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  300. Xiang C, Wang Y, Zhang H, Han F (2017) The role of endoplasmic reticulum stress in neurodegenerative disease. Apoptosis 22:1–26. https://doi.org/10.1007/s10495-016-1296-4

    Article  CAS  PubMed  Google Scholar 

  301. Xiong Z-G, Zhu X-M, Chu X-P, Minami M, Hey J, Wei W-L, MacDonald JF, Wemmie JA, Price MP, Welsh MJ et al (2004) Neuroprotection in ischemia: blocking calcium-permeable acid-sensing ion channels. Cell 118:687–698. https://doi.org/10.1016/j.cell.2004.08.026

    Article  CAS  PubMed  Google Scholar 

  302. Xu W, Qiu C, Gatz M, Pedersen NL, Johansson B, Fratiglioni L (2009) Mid- and late-life diabetes in relation to the risk of dementia: a population-based twin study. Diabetes 58:71–77. https://doi.org/10.2337/db08-0586

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  303. Yamada M, Kasagi F, Sasaki H, Masunari N, Mimori Y, Suzuki G (2003) Association between dementia and midlife risk factors: the radiation effects research foundation adult health study. J Am Geriatr Soc 51:410–414. https://doi.org/10.1046/j.1532-5415.2003.51117.x

    Article  PubMed  Google Scholar 

  304. Yang T, Sun Y, Lu Z, Leak RK, Zhang F (2017) The impact of cerebrovascular aging on vascular cognitive impairment and dementia. Ageing Res Rev 34:15–29. https://doi.org/10.1016/j.arr.2016.09.007

    Article  PubMed  Google Scholar 

  305. Yang Y, Jiang G, Zhang P, Fan J (2015) Programmed cell death and its role in inflammation. Mil Med Res 2:12–12. https://doi.org/10.1186/s40779-015-0039-0

    Article  PubMed  PubMed Central  Google Scholar 

  306. Yang Z, Zhang N, Shen H, Lin C, Lin L, Yuan B (2014) Microglial activation with reduction in autophagy limits white matter lesions and improves cognitive defects during cerebral hypoperfusion. Curr Neurovasc Res 11:223–229. https://doi.org/10.2174/1567202611666140520124407

    Article  CAS  PubMed  Google Scholar 

  307. Yarlagadda A, Alfson E, Clayton AH (2009) The blood brain barrier and the role of cytokines in neuropsychiatry. Psychiatry 6:18–18

    PubMed  PubMed Central  Google Scholar 

  308. Yassi N, Hilal S, Xia Y, Lim YY, Watson R, Kuijf H, Fowler C, Yates P, Maruff P, Martins R (2020) Influence of comorbidity of cerebrovascular disease and amyloid-β on Alzheimer’s disease. J Alzheimers Dis 73:897–907

    Article  CAS  PubMed  Google Scholar 

  309. Yenari MA, Kauppinen TM, Swanson RA (2010) Microglial activation in stroke: therapeutic targets. Neurother J Am Soc Exp NeuroTher 7:378–391. https://doi.org/10.1016/j.nurt.2010.07.005

    Article  CAS  Google Scholar 

  310. Yin F, Boveris A, Cadenas E (2014) Mitochondrial energy metabolism and redox signaling in brain aging and neurodegeneration. Antioxid Redox Signal 20:353–371. https://doi.org/10.1089/ars.2012.4774

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  311. Zhang LY, Pan J, Mamtilahun M, Zhu Y, Wang L, Venkatesh A, Shi R, Tu X, Jin K, Wang Y et al (2020) Microglia exacerbate white matter injury via complement C3/C3aR pathway after hypoperfusion. Theranostics 10:74–90. https://doi.org/10.7150/thno.35841

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  312. Zheng Y, Zhang J, Zhao Y, Zhang Y, Zhang X, Guan J, Liu Y, Fu J (2020) Curcumin protects against cognitive impairments in a rat model of chronic cerebral hypoperfusion combined with diabetes mellitus by suppressing neuroinflammation, apoptosis, and pyroptosis. Int Immunopharmacol 93:107422

    Article  CAS  PubMed  Google Scholar 

  313. Zhou Y, Zhang J, Wang L, Chen Y, Wan Y, He Y, Jiang L, Ma J, Liao R, Zhang X et al (2017) Interleukin-1β impedes oligodendrocyte progenitor cell recruitment and white matter repair following chronic cerebral hypoperfusion. Brain Behav Immun 60:93–105. https://doi.org/10.1016/j.bbi.2016.09.024

    Article  CAS  PubMed  Google Scholar 

  314. Zhu Y, Chai YL, Hilal S, Ikram MK, Venketasubramanian N, Wong B-S, Chen CP, Lai MKP (2017) Serum IL-8 is a marker of white-matter hyperintensities in patients with Alzheimer’s disease. Alzheimer’s Dement 7:41–47. https://doi.org/10.1016/j.dadm.2017.01.001

    Article  Google Scholar 

  315. Zhu Y, Hilal S, Chai YL, Ikram MK, Venketasubramanian N, Chen CP, Lai MKP (2018) Serum hepatocyte growth factor is associated with small vessel disease in Alzheimer’s dementia. Front Aging Neurosci 10:8–8. https://doi.org/10.3389/fnagi.2018.00008

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  316. Zuccalà G, Onder G, Pedone C, Carosella L, Pahor M, Bernabei R, Cocchi A (2001) Hypotension and cognitive impairment: selective association in patients with heart failure. Neurology 57:1986–1992. https://doi.org/10.1212/wnl.57.11.1986

    Article  PubMed  Google Scholar 

  317. Zuliani G, Ranzini M, Guerra G, Rossi L, Munari MR, Zurlo A, Volpato S, Atti A, Ble A, Fellin R (2007) Plasma cytokine profile in older subjects with late onset Alzheimer’s disease or vascular dementia. J Psychiatr Res 41:686–693. https://doi.org/10.1016/j.jpsychires.2006.02.008

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank the patients and their families for participating in the clinical studies cited in this review.

Funding

This work was supported by the National Medical Research Council of Singapore (MOH-000500-03, MOH-000707-01, MOH-001086-00, MOH-001086-00 to CPC, MKPL; NMRC-CBRG-0102/2016, NMRC/OFIRG/0036/2017 to TVA), Yong Loo Lin School of Medicine, National University of Singapore (Healthy Longevity Translational Research Programme HLTRP/2022/PS-01 to MKPL; Post-Doctoral Fellowship Award NUSMED/2021/PDF/05 to YLC), and La Trobe University (start-up grant to TVA).

Author information

Authors and Affiliations

Authors

Contributions

VR, TVA, CPC and MKPL developed the ideas for the review. DYF, DGJ, TMDS, GRD and CGS provided domain expertise. VR, YLC, LP and SS performed literature reviews. VR and YLC drafted the manuscript. All authors have read, edited and approved the submitted version of the manuscript.

Corresponding author

Correspondence to Mitchell K. P. Lai.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rajeev, V., Chai, Y.L., Poh, L. et al. Chronic cerebral hypoperfusion: a critical feature in unravelling the etiology of vascular cognitive impairment. acta neuropathol commun 11, 93 (2023). https://doi.org/10.1186/s40478-023-01590-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40478-023-01590-1

Keywords