Next Article in Journal
Myokines and Resistance Training: A Narrative Review
Previous Article in Journal
Potential Resistance of SARS-CoV-2 Main Protease (Mpro) against Protease Inhibitors: Lessons Learned from HIV-1 Protease
Previous Article in Special Issue
The Contribution of Small Vessel Disease to Neurodegeneration: Focus on Alzheimer’s Disease, Parkinson’s Disease and Multiple Sclerosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Small Vessel Disease: Ancient Description, Novel Biomarkers

Neurology Clinic, Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(7), 3508; https://doi.org/10.3390/ijms23073508
Submission received: 1 March 2022 / Revised: 20 March 2022 / Accepted: 21 March 2022 / Published: 23 March 2022
(This article belongs to the Special Issue Small Vessel Disease: New Perspectives on an Emerging Reality)

Abstract

:
Small vessel disease (SVD) is one of the most frequent pathological conditions which lead to dementia. Biochemical and neuroimaging might help correctly identify the clinical diagnosis of this relevant brain disease. The microvascular alterations which underlie SVD have common origins, similar cognitive outcomes, and common vascular risk factors. Nevertheless, the arteriolosclerosis process, which underlines SVD development, is based on different mechanisms, not all completely understood, which start from a chronic hypoperfusion state and pass through a chronic brain inflammatory condition, inducing a significant endothelium activation and a consequent tissue remodeling action. In a recent review, we focused on the pathophysiology of SVD, which is complex, involving genetic conditions and different co-morbidities (i.e., diabetes, chronic hypoxia condition, and obesity). Currently, many points still remain unclear and discordant. In this paper, we wanted to focus on new biomarkers, which can be the expression of the endothelial dysfunction, or of the oxidative damage, which could be employed as markers of disease progression or for future targets of therapies. Therefore, we described the altered response to the endothelium-derived nitric oxide-vasodilators (ENOV), prostacyclin, C-reactive proteins, and endothelium-derived hyperpolarizing factors (EDHF). At the same time, due to the concomitant endothelial activation and chronic neuroinflammatory status, we described hypoxia-endothelial-related markers, such as HIF 1 alpha, VEGFR2, and neuroglobin, and MMPs. We also described blood–brain barrier disruption biomarkers and imaging techniques, which can also describe perivascular spaces enlargement and dysfunction. More studies should be necessary, in order to implement these results and give them a clinical benefit.

1. Introduction

Small vessel disease (SVD) (also called cerebral small vessel disease, cSVD) relies on the deep brain’s small vessels alterations. Small vessels are univocally defined as small penetrating arteries, capillaries, and small veins. cSVD is strongly related to a chronic hypoperfusion condition, which predisposes the entire brain to hemorrhagic events, white (confluent or not) matter alterations, and lacunar events. SVD is the most important and common cause of all the vascular forms of dementia (up to 45%), but as previously underlined, it predisposes to a higher risk of vascular strokes (25–30% of cases) and 25–35% of all the lacunar events [1,2]. The other crucial common characteristic of SVD is that its pathological consequence could be represented by a silent lesion progression, which has its clinical confirmation in dramatic radiological imaging, without apparent, evident acute events. Thus, SVD is a clinical condition whose principal stigma is that the lesions may progress over time, for imprecise rules, and above all, with or without clinical consequences, in relationship with the extension and the confluency of the white matter alterations [3,4,5].
Generally, SVD clinical signs are concomitant psychological and behavioral sequelae, summarized by an essential executive function disruption and standard neuropsychological features (apathy and vascular depression) [1,2,3,4].
In sporadic cerebral SVD, aging, diabetes, chronic hypoxia, and hypertension are the most recognized clinical risk factors. Still, different hereditary forms of cerebral SVD have also been described [6]. Small arterioles show significant disruptions in both cases, easily described as arteriolosclerosis, lipohyalinosis, and severe endothelial disruption. Its principal consequence is a strong invalidation of the neurovascular coupling mechanisms and vessel tone dysregulation [7,8], and even venules are interested in the ongoing process [9]. With the arteriolosclerotic process, SVD is characterized by a substantial increment in cerebral amyloid angiopathy (CAA). This condition, which has been traditionally related to Alzheimer’s disease, is, on the contrary, quite frequent in the normal aging process, and it is dramatically evident in the SVD process. It is related to a consistent deposition of amyloid b-peptide (Ab) in the walls of the small arterioles, and it increments the consequences of altered neurovascular coupling in small parenchymal and leptomeningeal arterioles [9,10,11].
The principal consequence of arteriolosclerosis is the chronic hypoperfusive state, which induces a perpetual neuro-inflammation state, and gives rise to an essential endothelial activation. These conditions induce an overwhelming alteration of the oxidative response, which potentiates the basal inflammation status of the deep brain structure, expanding through different neural networks, principally the basal-forebrain ones [12].
In a recent review [12], we focused on the contribution of the complex and multifaceted “vascular damage” in developing small vessel dementia, starting from small vessel disease condition. We have written that the SVD is “an ongoing process, which begins with altered microvessels and pial arteries and ends in subcortical dementia; CBF regional selective decrease seems to be one of the critical factors for the progression from small vessel disease to small vessel disease-related dementia, together with proved altered response to inflammation, and oxidative stress” [12].
Neuroimaging is the main helpful diagnostic instrument for managing brain SVD. Therefore, the main findings in SVD are subcortical infarcts, lacunes, white matter hyperintensities (WMHs), prominent perivascular spaces (PVS), and cerebral microbleeds (CMBs) [13]. T2 or FLAIR MRI reports indicate confluent and symmetrical white matter hyperintensities [14,15,16,17,18,19,20,21,22,23] into the frontal and prefrontal-thalamus-basal forebrain networks [24,25,26,27,28,29]. Many instruments have been implemented to relate the number of lacunes, the extension and the amount of surface of white matter hyperintensities, and their relation to the subsequent cognitive and behavioral impairment [30,31,32]. The confluence between clinical, neuropsychological, and neuroimaging findings helps to converge for a correct diagnosis of the Vascular cognitive impairment, as stated in NINDS-AIREN criteria [33,34,35] and the DSM-V R (Fifth Edition-revised) [36,37,38,39]. VCI refers to an ample spectrum of vascular brain pathologies that contribute to cognitive impairment, ranging from mild and subjective cognitive decline to overt dementia [40,41].
The state of the art in the VCI field is an ongoing definition [42], and many terms have been employed, such as the descriptive ones of vascular cognitive disorder, subcortical vascular dementia, and mild and major vascular neurocognitive disorders (Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5) [36,37,38,39,43]. Others include vascular cognitive disorder (VCD), while subcortical VAD (sVAD) has been employed to define a circumscribed syndrome, related to small vessel disease [44,45,46]. However, it is well defined that the difference between VaD subtypes may depend on the anatomical distribution of the vascular insults [47]. Usually, small artery disease is more often associated with subcortical VaD than with cortical and cortical-subcortical VaD [47]. Executive dysfunctions and behavioral disorders (apathy and vascular depression, etc.) are the commonest findings [48].
Very recently, excellent studies emerged on the potential role of transcranial doppler findings in patients with white matter lesions, as possible markers for developing vascular dementia. A significant example is the demonstration that patients with white matter lesions, but without any other sign of cognitive impairment, showed a hemodynamic patter of cerebral hypoperfusion and enhanced vascular resistance, as a distinctive marker of a possible predictor factor of developing dementia [49]. Another significant result is the one obtained by a study using a transcranial Doppler, in which patients with ≥80% unilateral internal carotid artery stenosis with no history of stroke were recruited [50]; this study demonstrated that cognitive impairment correlated linearly with lower flow in the hemisphere fed by the occluded internal carotid artery, but only below a threshold of MFV = 45 cm/s. [50]
Finally, transcranial doppler studies have shown that it could delineate a profile of low perfusion and high vascular resistance in patients with a defined diagnosis of vascular depression [49].
In this paper, we wanted to address the contribution of the chronic inflammatory brain condition due to SVD and, starting from this situation, verify the possibility of finding new biomarkers of endothelial dysfunction, inflammation, and oxidative damage, which could be possible future targets of focused therapies.

2. Possible and Proved New Markers of Blood–Brain Barrier Leakage, Perivascular Enlargements, and Mitochondrial Alterations

SVD has the small vessels (pial and the small penetrating) and white matter as a significant definite target. Nevertheless, growing attention has been dedicated to disrupting perivascular spaces, astrocytic end-feet, capillaries, and veins. As a final point, the blood–brain barrier (BBB) has been addressed as another potential target of the intrigued mechanisms that underlie the small vessel brain pathology complex. BBB is not only a solid defensive barrier but acts as an active and specific player of active selection crossover, possessing cell-cell signaling with the end-feet of astrocytes and disclosure a potential role of maintaining efflux pumps [51,52,53,54,55]. Thus, the disruption of the BBB is proportionately increased by normal aging but progresses as a hallmark in different pathologies, i.e., multiple sclerosis or in primary inflammatory disease. Nevertheless, it is an expression of white matter inflammation, even due to chronic hypoperfusion, such as the one which occurs in small vessel disease [SVD], accomplishing the progression and the extension of the white matter sufferance, named as white matter hyperintensities (WMH) [56,57,58,59,60,61,62], the confluency of which is synonymous with SVD progression, leading to subcortical vascular dementia (sVAD) [12,62]. In AD-prone patients, BBB disruption has been signaled even in hippocampal degeneration, which occurs after a major stroke [61,62].
A dynamic contrast-enhanced MRI (DCE-MRI) [63] has been employed for in-vivo quantification of the pathological passage of plasma through BBB [64,65]. Moreover, apart from the BBB leakage, the possibility of estimating the vascular permeability-surface area product (PS) and the plasma volume fraction (VP) in a given region of interest has also been described [66,67]. The model suggested that PS increased with WMH severity, aging, and other vascular risk factors, and at the same time, a lower blood vP [65]. The most promising in-vivo demonstration is that BBB integrity is compromised in more severe WMH, even beyond visible lesions [63] (Insert Figure 1).
Even if we know that BBB is disrupted in SVD, we do not know the reasons for BBB leakage in this condition. The most disputed involvement is one of the pericytes. Pericytes are capillary mural cells that stabilize newly formed vessels and induce repair. When a pericyte-deficient adult mouse model has been employed [68], different transcriptional changes in brain endothelial cells have been mapped due to a defective pericyte contact at a single-cell level. In that conformation, endothelial cells, deprived of pericyte contacts, seem to exhibit a “venous-shifted molecular pattern,” and therefore lack any capillary specialization, and upregulate proteins which are typically expressed during developmental stages, such as the Fibroblast Growth Factor Binding Protein (Fgfbp1), or those expressed during pathological angiogenesis, such as Angiopoietin 2 (Angpt2). These aspects permit a possible cell proliferation, with a very flawed arteriolar BBB regulation system, and reduction of the angiogenesis process [68]. Fgfbp1 and Angpt2 levels could probably be crucial markers of BBB leakage during SVD. More studies will be necessary to prove that.
Perivascular spaces (PVS) have gained an essential role in SVD pathogenesis; they are no longer considered as virtual empty spaces, but as the most efficacious catabolites clearance system [12]; they are resident sites of perivascular macrophages, pial cells, mast cells, nerve fibers, and collagen fibers [69]. PVS are virtual spaces intimately connected to deep arterioles [70]. Even in these conditions, they act as a lymphatic net, defined as a glymphatic-perivascular territory [71].
Their malfunction, the hallmarks of which are the combined enlargement and widening, is the principal responsibility for perivascular accumulation of catabolites and toxic substances, which is determinant for enhancing ongoing neural damage until starvation [72,73]. The perivascular debris accumulation, together with the BBB leakage, potentiates and accelerates the perivascular inflammation, strongly favored by the stagnation-induced process and by medical conditions which influence it, such as hypertension and diabetes [74,75,76,77,78]. PVS enlargement is responsible for an altered cerebrovascular reactivity (CVR) [12], due to the extension of the constant inflammatory response [41] present as a constant marker in SVD, due to the chronic hypoperfusion state. The PVS is never an isolated situation, but it is accompanied by an altered BBB disruption and a significant perivascular inflammation [75,79,80,81,82]. More recently, new actors contribute with BBB leakage and PVS enlargement to help the progress of SVD [83,84,85,86], such as the oligodendrocyte precursor cells (OPCs), which generally help BBB stabilization [86,87] and the astrocytes, which exert their fundamental role as regulating the signal of neuro-vascular coupling [12]. Oligodendrocytes are the first victims of chronic models of chronic cerebral hypoperfusion (CCH), together with the precocious sufferance of the perineural space [88,89,90], and with a hyper-activation of microglia, firstly in the hippocampus [91,92], then in the thalamus, up to in the cortical neuronal population [93]. Secondary to oligodendrocytes, astrocyte death occurs in proportion to the chronic ischemia condition’s length and severity [94,95], due to the ongoing modifications of general and neuronal metabolic requests. Their death is a consequence of chronic hypoxia, but it worsens neuronal death due to a lack of functions, regulating the neurovascular coupling signal [96]. The process by which this occurs is that during the entire process of chronic ischemia, microglia retract its branches, with a consequent reduction of the length and strength of the microglial ramification, with a concomitant degeneration of the soma [97]. The frontal activation of microglia occurs in a two-step pattern: at the beginning, M1 activation upregulates TNF alpha, Il-23, IL-1beta, and Il12 production, which attack neurons, and directly contribute to their injury; only after M2 activation occurs can the reparation process can begin [98]. In the SVD, due to the chronic hypoxia-hypoperfusion condition [12], the passage through M1 towards M2 activation does not occur [98]. In SVD, there is a substantial augmentation of M1 activation, together with a heavy reduction of M2 promotion [99,100]. The brisk oligodendrocyte degeneration, associated with M1 activation, increases calcium currents and induces a severe apoptosis process. The calcium increases, and the severe apoptosis is accompanied by an augmentation of caspase-3 RNA and matrix-metalloprotease 2 (MMP-2) [101]. At the beginning of the SVD process, these markers reflect the temptation reparation process induced by a standard M1/M2 passage, as described above. Nevertheless, until the chronic inflammatory condition occurs in SVD ongoing development, there is an alteration of the M1/M2 passage, with a predominant M1 event; therefore, in SVD patients’ cerebrospinal fluid (CSF), there is a constant growth of oligodendrocyte-derived myelin sheath-like myelin lipid sulfatide (ODMSMS) and myelin essential protein (MBP) due to the massive oligodendrocytes death [102,103,104,105]. For similar reasons, markers of axonal damage, i.e., neurofilament light chain (NFL), together with CSF α-1 antitrypsin, tissue inhibitor of metalloproteinase-1 (TIMP-1), plasminogen activator inhibitor-1 (PAI-1), and apolipoprotein H (ApoH) have been found to increase very early in the CSF in SVD [106,107,108]. Finally, due to the BBB leakage, ultrastructural studies find that in older animals as well as in those affected by SVD, there are severe alterations of the capillary basement membrane of the deeper arterioles, inside the white matter, filling plasma proteins into vascular bagging and collagen deposition inside PVS, in a phenomenon described as microvascular fibrosis [55,98,109]. Many studies have testified that microvascular fibrosis and BBB splitting have a higher CSF/serum albumin (SA) ratio in patients with SVD [109]. Matrix remodeling pathway (TIMP-1 and matrix metalloproteinases) as an expression of endothelium disruption in SVD has been described [109] (Insert Table 1 here).

3. Markers of Endothelial Dysfunction

As previously described [12], there is a global endothelial altered function in SVD [110,111], which could be synthesized in an alteration of normal endothelial response to endothelium-derived nitric oxide-vasodilators (ENOV) [112], prostacyclin [113], C-reactive proteins [114], and endothelium-derived hyperpolarizing factors (EDHF) [115].
NO is rapidly removed in SVD for the mitochondrial alterations, with a consequent anti-oxidative response and consumed by peroxynitrite (O2 anions plus NO) [116]. However, it can also be reduced in its production, as it occurs in normal aging [117], in an accelerated way, in SVD, with a consistent down-regulation of endothelial NO synthase (eNOS). Moreover, in SVD, there is an evident dysfunction of the Rho-associated protein kinase (ROCK) [118] and the related ERM proteins (ezrin, radixin, and moesin), fundamental for barrier properties’ integrity [118,119,120] and their induction of the downregulation of the vascular endothelium cadherins (VE-cadherins) [121].
In diabetes, where SVD is a constant presentation form with a crucial endothelial hyper-permeability, a concomitant increase in arteriolar deposition of advanced glycation end products has been observed, which helps and maintains the increase in endothelial permeability through Rho activation and an upregulation of the vascular endothelial growth factor (VEGF) [122,123].
The superimposition of BBB disruption, endothelial dysfunction, and microvascular fibrosis causes a substantial permeability alteration, with albumin extravasation; the increased CSF/plasma albumin ration is a proven witness of a severe progression of confluency of white matter lesions in SVD [124,125,126,127], together with albuminuria (even if not well-accepted) [128,129,130,131].
Other important markers of endothelial altered activation [12,132,133,134] in SVD are intercellular adhesion molecule-1 (ICAM-1), which has been considered as a generic expression of white matter progression [95], soluble thrombomodulin (sTM), interleukin-6 (IL-6), plasminogen activator inhibitor-1 (PAI-1), and von Willebrand factor [129,130,131,132,133,134]. Others, such as HIF 1 alpha, VEGFR2, and neuroglobin, are more evident when the confluency of different WMH becomes constant in different models [135,136].

4. Markers of Oxidative Damages in SVD

Reactive oxygen species (ROS) is an umbrella term for many ordinary derivatives of molecular oxygen, and their accumulation leads to a complex phenomenon called oxidative distress. There are two species, hydrogen peroxide (H2O2) and the superoxide anion radical (O2), which are key redox signaling agents generated under the control of growth factors and cytokines by more than 40 enzymes, prominently including nicotinamide adenine dinucleotide phosphate (NADPH) oxidases [12] and the mitochondrial electron transport chain [126]. When mitochondrial cells usually function, the active process of oxidative phosphorylation converts oxygen to superoxide by oxidase enzymes, and superoxide can be transformed by superoxide dismutase (SOD) or to non-radical hydrogen peroxide [126,136,137], i.e., from glutathione peroxidase (Gpx), or when catalase enzymatically metabolizes hydrogen peroxide to water and oxygen [136].
Chronic cerebral conditions of constant hypoxia are the principal inductors of the uncontrolled production of ROS [138,139].
NADPH oxidase activity and mitochondrial are significantly higher in cerebral arteries when compared with systemic arteries in blood vessels from healthy animals (mouse, rat, pig, and rabbit) [140,141]. Thus, brain vessels are one of the most prominent productions of ROS, suggesting that there could be fundamental ROS-dependent signaling in cerebral arteries, which might be indispensable for vasoactive regulation properties.
Thus, the accumulation of ROS species, associated with mitochondrial dysfunction, BBB disruption, and chronic inflammatory status are three conditions in SVD and are proportionate to WMH extension. They lead to an altered endothelial further altered activation, which is reflected in a decoupling of the neurovascular coupling system, with significant sub-cortical and cortical signal alteration, with consequent reflex in oligodendrocytes astrocytes and finally to neurons [12,142]. An active role of flow-dependent responses in rat cerebral arteries has been recently demonstrated in vivo, directly exerted by the NADPH-oxidase reactions [143]. Specifically, Nox2-NADPH oxidase dysfunction is related to the propagation of the ischemic brain injury, derived by the occlusion of larger pial arteries; Nox2/NOx2 knock-out mice, in the same condition, show the minor extension of brain injury after an ischemic infarct [144].
The induced alterations of mitochondrial DNA by ROS attacks and chronic ischemic conditions are some of the most critical contributors to neuronal aging and degeneration, either considering oxidative damage as a promoter or as a consequence of it [145,146,147].
The decline of mitochondrial functioning has been largely implicated in the aging process and is characterized by a reduced density of mitochondria and reduced mitogenesis [148,149,150,151,152]. Such changes, which originate as replication errors, accumulate in postmitotic tissues during aging, leading to increased proportions of impaired mitochondria [152]. In the aging brain, there has been a sufficient demonstration of impairment of synaptic mitochondria leading to impaired neurotransmission and cognitive failure [149,150,151,152,153,154,155]. Precocious forms of small vessel disease, leading to vascular dementia, have been described in specific mitochondrial point mutation [156]. Other mitochondrial mutation phenotypes have been described as pure brain involvement, including fluctuating encephalopathy, seizures, dementia, migraine, stroke-like episodes, ataxia, and spasticity [149,153,154,155]. Growing attention should be paid to mitochondrial DNA mutations for brain pathologies, in order to gain more robust data on their possible relevance, and their correlation with postmortem neuropathologic features, to advance our understanding [156,157,158,159,160,161].
Oxidative stress potentiates the disorders of the endothelium-dependent NO signaling [162,163]. Uncoupling endothelial NO synthase (eNOS) (i.e., in relation with lower levels of tetrahydrobiopterin) switches the production of NO to that of superoxide, causing an overwhelming potentiation of ROS production, accelerating the oxidative stress, lowering the NO anti-inflammatory properties [164,165], and reducing NO modulation of Rho-kinase activity, inhibiting vascular tone control [166]. Rho-kinase, as a counterpart, influences mRNA-stability of eNOS [167].
The induction of oxidative stress is one of the most important promoters of pathological angiogenesis, by lipid oxygenation, thickening the blood vessel walls [168,169]. Moreover, the ApoE4 allele and the AD process seem to be involved in promoting vascular alterations independently of other recognized factors, i.e., age, diabetes, hypertension, and obesity, etc. However, it is supposed to worsen the confluency of WMH, probably somehow linked to ROS augmentation, without any other positive data [170,171,172].

5. Inflammation and SVD

As above written, neuroinflammation is a common finding in SVD models; it is tightly related to chronic hypoperfusion condition and defined as located hypoxia condition, the common finding of SVD. The pivotal role of neuroinflammation in SVD could accelerate the lipid peroxidation precipitation of the redox system and promote a more robust activation of M1 than M2 [173].
It has been demonstrated that NO-related metabolite, citrulline, and dimethylarginine (DMA) concentrations were significantly higher in patients with strategic infarcts [174]. Arginine depletion was an independent predictor of VaD [174]. S100B (calcium-binding protein B) is a protein that stimulates the expression of pro-inflammatory cytokines. SomIt has been described to have a significant correlation between S100B/asymmetric dimethylarginine levels and cognitive decline in patients with leukoaraiosis [175,176].
Homocysteine could be a potential marker of neuroinflammation inside SVD, promoting the increase in TNF-alpha and IL1-beta, upregulating the transcriptional fibroblast growth factor-2, IL-6, and IL-8, [177,178], and enhancing the VEGF/ERK1/2 signaling pathway [179,180], which can be seen frequently in the atherosclerosis process. Homocysteine is directly linked to the B-inflammatory pathway through a direct upregulation of pyruvate kinase muscle isoenzyme 2 (PKM-2), B-mediated, which mainly promotes the inflammatory basis of atherosclerosis cascade [181,182].
Homocysteine accumulation promotes an increase in the endoplasmic reticulum (ER) stress, upregulating metalloproteinases-9 (MMP-9), and inducing apoptosis [183]. Definitively, the accumulation of homocysteine in animal models enhanced the expression of the AGEs or vascular cell adhesion molecule [184] and MMP-9 [185]. The inflammation cascade could be mediated by the effects on smooth muscle cells rather than on the endothelium alterations [186,187].
Chronic inflammation and oxidative stress have been suggested as concurrent mechanisms of SVD. A possible link between accumulation products (i.e., homocysteine) and other markers could be the circulating metalloproteinases (MMPs) and the tissue inhibitors of metalloproteinases (TIMPs) [188].
MMPs, some of the Ca2+-Zn endopeptidase, have been described as having six different properties: collagenase, gelatinases, stromelysins, matrilysin, membrane-specific metalloproteinases, and no other specified. Their specific role inside the brain is complex and multifaceted; it begins with the neuronal networks remodeling throughout the integrity of the BBB [189]. MMP remains inside the brain, probably in inactivate form, and is active only under special conditions, such as chronic hypoperfusion or chronic inflammatory status. The significant components are MMP2 and MMP14, which are present specifically inside astrocytes, whereas microglia present the MMP-3 and the MMP9, which, by definition, are called inflammatory metalloproteases. Their expression is more severe in acute damage and gradually decreases in the reparation phases. They can be found near the damaged areas and in the propinquity vessel-related areas [190].
There are four possible mechanisms which have been related to MMP involvement in SVD and, in general, in the neuroinflammation process. The most obvious and well-studied MMP directly activates signaling cytokines, cell-receptors, and adhesion molecules. There are essential works that testify that, even directly, MT4-MMP upregulates a TNF-alpha convertase, and is able to activate TNF-alpha, in its soluble and active [191,192]. Secondly, there are many pieces of evidence in different clinical cases (neurological bacterial infection and PD, etc.), in which there is a direct activation, probably mediated by lipopolysaccharides, calcium currents and other apoptotic signals, and alpha-synuclein deposits, which activate MMP-3 into the interstitial brain fluid, and there, it triggers M1 activation, with a consequent (and above-described) M1 activation [193,194].
Thirdly, MMP seems to be tightly involved in the so-called Fas-FasL system. This system has been known as an inducer of extrinsic cell death responsible for cell-mediated cytotoxicity and peripheral immune regulation. MMP might improve the FAS system, probably through an intrinsic possibility of modulating chloride channel activity, inducing and promoting glutamate excitotoxicity currents, or altering the interactions between neuronal cells and extracellular matrix compounds [195].
Finally, the MMPs participate in many digestive processes at the BBB, particularly the tight junctions and the basement membrane. It has been proposed that MMPs digest tight junctions and basement membrane proteins, thus contributing to BBB leakage [196]. The increased activity of MMP, tightly associated with a higher permeability at the BBB, has been demonstrated in vivo during the reperfusion process through an increase in MMP-2 and MMP-9 mRNA activity [197]. The induction of BBB leakage has, as an indirect effect, an increment in the vasogenic edema, inside the WM, with a drastic increment in vascular demyelination process (Insert Figure 2).
These data have been evoked in animal models and rare human models, and there is a substantial lack of information, i.e., on the possible relationship between MMP levels and extension and repairing of stroke lesions [198,199].
Nevertheless, some interesting points shed some light on the topic: increased confluency of WMH could be related to higher levels of TIMP-4, after three months of a primary stroke [199].
In a recent study, Arba et al. showed that increasing the grade of SVD sustains higher levels of TIMP-4 and supports the involvement of TIMP-4 in the pathologic process of SVD; they studied a population of an ischemic stroke patient, reporting that brain atrophy was associated with baseline TIMP-4 levels and leukoaraiosis was associated with 90-day TIMP-4 levels. A global SVD score, expressed as a combined product of leukoaraiosis, lacunes, and brain atrophy, was associated with TIMP-4 levels at 90 days with a dose-response effect [199].
Increased levels of MMP have been associated with severe white matter alterations and a cognitive profile that resembles sVAD [200,201]. In particular, a positive relationship between MMP2 lower levels has been found, together with an increase in albumin index in CSF of SVD patients, as above written [202,203,204].
Due to lipohyalinosis substitution of smooth muscle cells in arterioles (as described in 12), there is an inverse correlation between TIMP-4 elevated levels (only in animal models) and reduction of lipohyalinosis and collagen bagging through an undescribed and uncertain mechanism [205,206,207].
All these aspects accounted for, MMPs and tissue inhibitors of metalloproteinases-1 (TIMP-1) could be promising SVD biomarkers [208,209].

6. Potential Future Therapies Approach

Different approaches can be employed to offer potential treatment for VCI, at the moment these are only symptomatic; many data have been obtained from cholinesterase inhibitors and memantine [210].
Potential treatment strategies for brain SVD might include those that target antioxidant effects for the endothelium of small cerebral vessels and the BBB. Due to the major decrease in NO bioavailability in SVD, NO donors could help release the functioning endothelium of small vessel disease, limited by their susceptibility to tolerance development. The apparent strategy, in the same manner as the administration of potent antioxidants such as Vitamins C and E, has shown to be beneficial for vascular function in several experimental and small clinical trials [211].
Disappointingly, the results of large clinical trials of antioxidant supplementation have largely failed to show any benefit. The ROS scavenger tempol is cell-permeable and has been used in experimental studies, as well as edaravone (O2-scavenger). The ROS scavenger tempol is cell-permeable and has been used in experimental studies, as well as edaravone (O2-scavenger). Problems derived from NADPH oxidase activity, particularly its primary contributor, Nox2. It can be argued that prolonged selective therapies could help prevent brain SVD but invariably lead to an immunosuppression condition and many other side effects derived by other different Nox oxidases [137,145,212].
Notably, three of the most influential and frequently prescribed classes of drugs for the treatment of vascular risk factors, which have been shown to inhibit NADPH oxidases, reducing oxidative stress, are the Angiotensin-converting-enzyme inhibitors (ACE inhibitors), Angiotensin II receptor type 1 (AT 1) antagonists, and the statins [3,4]. There are no impressive studies on these drugs as primary NADPH inhibitors, rather than their well-known function per se.
Many other trials have been conducted and are still ongoing [213].
Phenolic acids (or phenolcarboxylic acids) are aromatic acid compounds containing a phenolic ring and an organic carboxylic acid function [214]. Among the most studied molecules belonging to this group, caffeic, chlorogenic, o-coumaric, p-coumaric, m-coumaric, ferulic, and cinnamic acids are the most commonly consumed in the human diet, being contained in coffee [215], together with gallic, p-hydroxybenzoic, vanillic, syringic, and protocatechuic acids. They can be found in bran, grain brown rice, olive oil, tea, cherries, plums, gooseberries, and red wine [216]. These substances have been studied among middle-aged adults, showing a benefit of their intake in different cognitive domains [217,218,219,220,221,222,223,224].
In the same way, rosmarinic acid induced a promotion of oxidative stress response and a reduced lipid-peroxidation [225], but also reduced the gene expression of inducible nitric oxide synthase [225,226], and promoted neuroprotection, reducing matrix metallopeptidase 2 (MMP2), and IL-1 beta [225,226,227]. Myrtenal has been recently employed as a multi-property substance (anti-inflammatory and anti-oxidant) [228] but results are only promising.
Apart from physical aerobic activity and avoiding vascular risk factors (smoking, high quantities of carbohydrates, and alcohol consumption, etc.), even external stimuli have been applied in studies; in order to implement cognitive abilities on vascular deterioration, transcranial magnetic stimulation has been studied, which it is still under debate, because its activity on the dorsal striatum with the consequential increase in dopamine release may contribute to the clinical and neurophysiological outcome in vascular depression and vascular cognitive impairment [229] (Insert Scheme 1).

7. Conclusions

In the last few decades, the concept of vascular contributions to cognitive impairment and dementia has been emphasized. Cerebral small vessel disease is a common neurocognitive disorder and source of disability. Pathophysiology of cSVD is complex, involving multiple pathways, as described before. Several risk factors, including genetic, co-morbid complications, and environmental factors, contribute to the pathogenesis or exacerbate the complications. Inflammation, chronic hypoperfusion, oxidative damage, glymphatic alterations, and BBB disruption might be potential contributors to the pathogenesis of this complex phenomenon. MMPs, ROS, and other reactive factors trigger inflammatory responses, leading to the abnormalities in small vessels and endothelium dysfunction associated with CSVD.
This study has several limits: Although comprehensive, the approach used in the examined investigations in the attempt to disentangle the complex pathomechanisms of VCI has a number of caveats and potential criticisms. So far in our study, we have tried to have the most homogenous definition, but otherwise, just examining different animal models could represent not a constant level of clinical reversibility. Therefore, the available results on a relatively small sample size might not be confirmed on larger populations, although most of them were obtained from homogeneous samples.
Another limitation is that the correlation between different techniques and the anatomical distribution and severity of vascular lesions has been rarely systematically investigated; therefore, without the contribution of advanced imaging, blood samples, cerebrospinal fluid, laboratory models, or the combination of techniques, the conclusions that can be reached cannot be sufficiently powerful.
Finally, results do not usually provide specific clinical information, although they are sensitive to the “global weight” of several biochemical pathways and neurotransmitter activities. Consequently, a panel of changes, rather than a single marker of disease, should be considered.
More detailed investigations are required to understand the pathophysiology of SVD. Several fluid biomarkers that might be used in diagnostic settings have been identified. Thus, currently, there is little value in blood tests. CSF biomarkers may help physicians separate vascular and neurodegenerative causes based on BBB disruption and extracellular matrix breakdown. Alongside the need for a correct diagnosis of the disease, biomarkers could be valuable tools to monitor the progression of the disease itself and the possible response to treatment. In this work, we have tried to underline the importance of the inflammatory response in disease pathogenesis. Much further work needs to be conducted along with these positions. The search for an optimal panel of biomarkers with high sensitivity and specificity will provide the crucial tools to enhance success in identifying valid biomarkers in SVD. A combination of biochemical and imaging markers and psychometrics will be necessary to improve the diagnostic accuracy progression of the pathology and finally to monitor response to possible treatment. We believe that the contribution of inflammation on SVD is significant and should be further studied to identify new therapeutic possibilities.

Author Contributions

Conceptualization R.M. and P.C.; methodology, R.M.; software, R.M. and P.C.; formal analysis, R.M. and P.C.; investigation, R.M.; data curation R.M. and P.C.; writing—original draft preparation, R.M. and P.C.; writing—review and editing, R.M.; visualization, R.M.; supervision, R.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AbAmyloid b-peptide
ADAlzheimer′s Disease
Angpt2Angiopoietin 2
ApoHApolipoprotein H
BBBBlood–-brain barrier
CAACerebral amyloid angiopathy
CBFCerebral blood flow
CCHChronic cerebral hypoperfusion
CNS Central Nervous System
COXCyclooxygenase
CSFCerebrospinal fluid
cSVD Cerebral small vessel disease
CVRCerebrovascular reactivity
ECEndothelial cells
EDHFEndothelium-derived hyperpolarizing factors
eNOSEndothelial NO synthase
ENOVEndothelium-derived nitric oxide-vasodilators
Fgfbp1Fibroblast growth factor binding protein
GABAGamma-aminobutyric acid
GPxGlutathione Peroxidase
GSHGlutathione
ICAM-1Intercellular adhesion molecule-1
ILInterleukin
ISFInterstitial fluid
MBPMyelin basic protein
MMPs Matrix metalloproteinase
NADPHNicotinamide adenine dinucleotide phosphate
NfLNeurofilament light chain
NONitric oxide
ODMSMSOligodendrocyte-derived myelin sheath-like myelin lipid sulfatide
OPCsOligodendrocyte precursor cells
PAI-1Plasminogen activator inhibitor-1
PSPermeability-surface area product
PVSPerivascular spaces
ROCK Rho-associated protein kinase
ROSReactive oxygen species
SASerum albumin
SVDSmall vessel disease
SODSuper Oxide Dismutase
sTMSoluble thrombomodulin
TIMPsTissue inhibitors of metalloproteinases
TNF-αTumor necrosis factor-α
VCIVascular cognitive impairment
VE-cadherins Vascular endothelium cadherins
VEGF Vascular Vascular endothelial growth factor
vPPlasma volume fraction
WMHWhite matter hyperintensities

References

  1. Hakim, A.M. Small Vessel Disease. Front. Neurol. 2019, 10, 1020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Uiterwijk, R.; Van Oostenbrugge, R.J.; Huijts, M.; De Leeuw, P.W.; Kroon, A.A.; Staals, J. Total Cerebral Small Vessel Disease MRI Score Is Associated with Cognitive Decline in Executive Function in Patients with Hypertension. Front. Aging Neurosci. 2016, 8, 301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Taylor, W.D.; Aizenstein, H.J.; Alexopoulos, G.S. The vascular depression hypothesis: Mechanisms linking vascular disease with depression. Mol. Psychiatry 2013, 18, 963–974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Pinter, D.; Ritchie, S.J.; Doubal, F.; Gattringer, T.; Morris, Z.; Bastin, M.; Hernández, M.D.C.V.; Royle, N.A.; Corley, J.; Maniega, S.M.; et al. Impact of small vessel disease in the brain on gait and balance. Sci. Rep. 2017, 7, 41637. [Google Scholar] [CrossRef] [PubMed]
  5. Wardlaw, J.M.; Smith, E.F.; Biessels, G.J.; Cordonnier, C.; Fazekas, F.; Frayne, R.; Lindley, R.I.; O’Brien, J.T.; Barkhof, F.; Benavente, O.R.; et al. Neuroimaging standards for research into small vessel disease and its contribution to ageing and neurodegeneration. Lancet Neurol. 2013, 12, 822–832. [Google Scholar] [CrossRef] [Green Version]
  6. Haffner, C.; Malik, R.; Dichgans, M. Genetic factors in cerebral small vessel disease and their impact on stroke and dementia. J. Cereb. Blood Flow Metab. 2016, 36, 158–171. [Google Scholar] [CrossRef] [Green Version]
  7. Moody, D.M.; Brown, W.R.; Challa, V.R.; Anderson, R.L. Periventricular venous collagenosis: Association with leukoaraiosis. Radiology 1995, 194, 469–476. [Google Scholar] [CrossRef] [PubMed]
  8. Smith, E.E.; Vijayappa, M.; Lima, F.; Delgado, P.; Wendell, L.; Rosand, J.; Greenberg, S.M. Impaired visual evoked flow velocity response in cerebral amyloid angiopathy. Neurology 2008, 71, 1424–1430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Park, L.; Koizumi, K.; El Jamal, S.; Zhou, P.; Previti, M.L.; Van Nostrand, W.E.; Carlson, G.; Iadecola, C. Age-Dependent Neurovascular Dysfunction and Damage in a Mouse Model of Cerebral Amyloid Angiopathy. Stroke 2014, 45, 1815–1821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Staals, J.; Booth, T.; Morris, Z.; Bastin, M.E.; Gow, A.J.; Corley, J.; Redmond, P.; Starr, J.M.; Deary, I.; Wardlaw, J.M. Total MRI load of cerebral small vessel disease and cognitive ability in older people. Neurobiol. Aging 2015, 36, 2806–2811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Staals, J.; Makin, S.D.; Doubal, F.N.; Dennis, M.S.; Wardlaw, J.M. Stroke subtype, vascular risk factors, and total MRI brain small-vessel disease burden. Neurology 2014, 83, 1228–1234. [Google Scholar] [CrossRef] [PubMed]
  12. Moretti, R.; Caruso, P. Small Vessel Disease-Related Dementia: An Invalid Neurovascular Coupling? Int. J. Mol. Sci. 2020, 21, 1095. [Google Scholar] [CrossRef] [Green Version]
  13. Tonet, E.; Pompei, G.; Faragasso, E.; Cossu, A.; Pavasini, R.; Passarini, G.; Tebaldi, M.; Campo, G. Coronary Microvascular Dysfunction: PET, CMR and CT Assessment. J. Clin. Med. 2021, 10, 1848. [Google Scholar] [CrossRef]
  14. Rosenberg, G.A.; Wallin, A.; Wardlaw, J.M.; Markus, H.S.; Montaner, J.; Wolfson, L.; Iadecola, C.; Zlokovic, B.V.; Joutel, A.; Dichgans, M.; et al. Consensus statement for diagnosis of subcortical small vessel disease. J. Cereb. Blood Flow Metab. 2016, 36, 6–25. [Google Scholar] [CrossRef] [Green Version]
  15. Patel, B.; Markus, H.S. Magnetic Resonance Imaging in Cerebral Small Vessel Disease and its Use as a Surrogate Disease Marker. Int. J. Stroke 2011, 6, 47–59. [Google Scholar] [CrossRef]
  16. Erkinjuntti, T.; Inzitari, D.; Pantoni, L.; Wallin, A.; Scheltens, P.; Rockwood, K.; Roman, G.C.; Chui, H.; Desmond, D.W. Resaerch criteria for subcortical vascular dementia in clinical trials. J. Neur. Transm. Suppl. 2000, 59, 23–30. [Google Scholar]
  17. O’Donnell, M.J.; Xavier, D.; Liu, L.; Zhang, H.; Chin, S.L.; Rao-Melacini, P.; Rangarajan, S.; Islam, S.; Pais, P.; McQueen, M.J.; et al. Risk factors for ischaemic and intracerebral haemorrhagic stroke in 22 countries (the INTERSTROKE study): A case-control study. Lancet 2010, 376, 112–123. [Google Scholar] [CrossRef]
  18. Potter, G.M.; Marlborough, F.J.; Wardlaw, J.M. Wide variation in definition, detection and description of lacunar lesions on imaging. Stroke 2011, 42, 359–366. [Google Scholar] [CrossRef] [PubMed]
  19. Shi, Y.; Wardlaw, J.M. Update on cerebral small vessel disease. A dynamic whole-brain disease. Stroke Vasc. Neurol. 2016, 1, e000035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Wardlaw, J.M.; Doubal, F.; Armitage, P.; Msc, F.C.; Carpenter, T.; Maniega, S.M.; Farrall, A.; Sudlow, C.; Dennis, M.; Dhillon, B. Lacunar stroke is associated with diffuse blood-brain barrier dysfunction. Ann. Neurol. 2009, 65, 194–202. [Google Scholar] [CrossRef]
  21. Englund, E. White matter pathology of vascular dementia. In Vascular Dementia; Chui, E., Ed.; M. Dunitz: London, UK, 2004; pp. 117–130. [Google Scholar]
  22. Englund, E.A.; Person, B. Correlations between histopathologic white matter changes and proton MR relaxation times in dementia. Alzheimer Dis. Assoc. Disord. 1987, 1, 156–170. [Google Scholar] [CrossRef] [PubMed]
  23. Smallwood, A.; Oulhaj, A.; Joachim, C.; Christie, S.; Sloan, C.; Smith, A.D.; Esiri, M. Cerebral subcortical small vessel disease and its relation to cognition in elderly subjects: A pathological study in the Oxford Project to Investigate Memory and Ageing (OPTIMA) cohort. Neuropathol. Appl. Neurobiol. 2012, 38, 337–343. [Google Scholar] [CrossRef] [PubMed]
  24. Gold, G.; Kovari, E.; Herrmann, F.R.; Canuto, A.; Hof, P.R.; Michel, J.P.; Bouras, C.; Giannakopoulos, P. Cognitive consequences of thalamic, basal ganglia, and deep white matter lacunes in brain aging and dementia. Stroke 2005, 36, 1184–1188. [Google Scholar] [CrossRef]
  25. Klassen, A.C.; Sung, J.H.; Stadlan, E.M. Histological changes in cerebral arteries with increasing age. J. Neuropathol. Exp. Neurol. 1968, 27, 607–623. [Google Scholar] [CrossRef]
  26. Cummings, J.L. Frontal-subcortical circuits and human behavior. Arch. Neurol. 1993, 50, 873–880. [Google Scholar] [CrossRef]
  27. Mega, M.S.; Cummings, J.L. Frontal-subcortical circuits and neuropsychiatric disorders. J. Neuropsychiatry Clin. Neurosci. 1994, 6, 358–370. [Google Scholar]
  28. Tak, S.; Yoon, S.J.; Jang, J.; Yoo, K.; Jeong, Y.; Ye, J.C. Quantitative analysis of hemodynamic and metabolic changes in subcortical vascular dementia using simultaneous near-infrared spectroscopy and FMRI measurements. Neuroimage 2011, 55, 176–184. [Google Scholar] [CrossRef] [PubMed]
  29. McKhann, G.; Drachman, D.; Folstein, M.; Katzman, R.; Price, D.; Stadlan, E.M. Clinical diagnosis of Alzheimer’s disease: Report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology 1984, 34, 939–944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Kramer, J.H.; Reed, B.R.; Mungas, D.; Weiner, M.W.; Chui, H. Executive dysfunction in subcortical ischaemic vascular disease. J. Neurol. Neurosurg. Psychiatr. 2002, 72, 217–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Burton, E.; Ballard, C.; Stephens, S.; Kenny, R.A.; Kalaria, R.; Barber, R.; OʹBrien, J. Hyperintensities and fronto-subcortical atrophy on MRI are substrates of mild cognitive deficits after stroke. Dement. Geriatr. Cogn. Disord. 2003, 16, 113–118. [Google Scholar] [CrossRef] [PubMed]
  32. Tullberg, M.; Fletcher, E.; DeCarli, C.; Mungas, D.; Reed, B.R.; Harvey, D.J.; Weiner, M.W.; Chui, H.C.; Jagust, W.J. White matter lesions impair frontal lobe function regardless of their location. Neurology 2004, 63, 246–253. [Google Scholar] [CrossRef] [PubMed]
  33. Román, G.C.; Tatemichi, T.K.; Erkinjuntti, T.; Cummings, J.L.; Masdeu, J.C.; Garcia, J.H.; Amaducci, L.; Orgogozo, J.M.; Brun, A.; Hofman, A. Vascular dementia: Diagnostic criteria for research studies. Report of the NINDS-AIREN International Workshop. Neurology 1993, 43, 250–260. [Google Scholar] [CrossRef] [PubMed]
  34. Chui, H.C.; Victoroff, J.I.; MArgolin, D.; Jagust, W.; Shankle, R.; Katzman, R. Criteria for the diagnosis of ischemic vascular dementia proposed by the state of California Alzheimer’s Disease Diagnostic and Treatment Centers. Neurology 1992, 42, 473–480. [Google Scholar] [CrossRef]
  35. Kim, G.H.; Lee, J.H.; Seo, S.W.; Ye, B.S.; Cho, H.; Kim, H.J.; Noh, Y.; Yoon, C.W.; Chin, J.H.; Oh, S.J.; et al. Seoul criteria for PIB(-) subcortical vascular dementia based on clinical and MRI variables. Neurology 2014, 82, 1529–1535. [Google Scholar] [CrossRef]
  36. Fazekas, F.; Chawluk, J.B.; Alavi, A.; Hurtig, H.I.; Zimmermann, R.A. MR signal abnormalities at 1.5 T in Alzheimer’s dementia and normal aging. Am. J. Roentgenol. 1987, 149, 351–356. [Google Scholar] [CrossRef] [Green Version]
  37. Cleutjens, F.A.H.M.; Ponds, R.W.H.M.; Spruit, M.A.; Burgmans, S.; Jacobs, H.I.L.; Gronenchield, H.B.M.; Stalls, J.; Franssen, F.M.E.; Dijkstra, J.B.; Vanfleteren, L.E.G.M.; et al. The relationship between cerebral small vessel disease, hippocampal volume and cognitive functioning in patients with COPD: An MRI study. Front. Aging Neurosci. 2017, 9, 88. [Google Scholar] [CrossRef]
  38. Scheltens, P.; Barkhof, F.; Leys, D.; Pruvo, J.P.; Nauta, J.J.; Vermersch, P.; Steinling, M.; Valk, J. A semiquantative rating scale for the assessment of signal hyperintensities on magnetic resonance imaging. J. Neurol. Sci. 1993, 114, 7–12. [Google Scholar] [CrossRef]
  39. Kim, K.W.; MacFall, J.R.; Payne, M.E. Classification of white matter lesions on magnetic resonance imaging in elderly persons. Biol. Psychiatry 2008, 64, 273–280. [Google Scholar] [CrossRef] [Green Version]
  40. O’Brien, J.T.; Erkinjuntti, T.; Reisberg, B.; Roman, G.; Sawada, T.; Pantoni, L.; Bowler, J.V.; Ballard, C.; DeCarli, C.; Gorelick, P.B.; et al. Vascular cognitive impairment. Lancet Neurol. 2003, 2, 89–98. [Google Scholar] [CrossRef]
  41. Erkinjunnti, T.; Gauthier, S. Diagnosing vascular cognitive impairment and dementia. In Concepts and Controversies in Vascular Cognitive Impairment in Clinical Practice; Wahlund, L.O., Erkinjunnti, T., Gauthier, S., Eds.; Cambridge University Press: Cambridge, UK, 2009; pp. 3–9. [Google Scholar]
  42. Van der Flier, W.M.; Skoog, I.; Schneider, J.A.; Pantoni, L.; Mok, V.; Chen, C.L.H.; Scheltens, P. Vascular cognitive impairment. Nat. Rev. Dis. Primers 2018, 4, 18003. [Google Scholar] [CrossRef]
  43. Skrobot, O.A.; O’Brien, J.; Black, S.; Chen, C.; DeCarli, C.; Erkinjuntti, T.; Ford, G.A.; Kalaria, R.N.; Pantoni, L.; Pasquier, F.; et al. The Vascular Impairment of Cognition Classification Consensus Study. Alzheimers Dement. 2017, 13, 624–633. [Google Scholar] [CrossRef] [Green Version]
  44. Thal, D.R.; Grinberg, L.T.; Attems, J. Vascular dementia: Different forms of vessel disorders contribute to the development of dementia in the elderly brain. Exp. Gerontol. 2012, 47, 816–824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Pantoni, L. Cerebral small vessel disease: From pathogenesis and clinical characteristics to therapeutic challenges. Lancet Neurol. 2010, 9, 689–701. [Google Scholar] [CrossRef]
  46. Bowler, J.V. Vascular cognitive impairment. J. Neurol. Neurosurg. Psychiatr. 2005, 76, 35–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Caruso, P.; Signori, R.; Moretti, R. Small vessel disease to subcortical dementia: A dynamic model, which interfaces aging, cholinergic dysregulation and the neurovascular unit. Vasc. Health Risk Manag. 2019, 15, 259–281. [Google Scholar] [CrossRef]
  48. Jellinger, K.A. Pathomechanisms of Vascular Depression in Older Adults. Int. J. Mol. Sci. 2021, 23, 308. [Google Scholar] [CrossRef] [PubMed]
  49. Puglisi, V.; Bramanti, A.; Lanza, G.; Cantone, M.; Vinciguerra, L.; Pennisi, M.; Bonanno, L.; Pennisi, G.; Bella, R. Impaired Cerebral Haemodynamics in Vascular Depression: Insights from Transcranial Doppler Ultrasonography. Front. Psychiatry 2018, 9, 316. [Google Scholar] [CrossRef] [Green Version]
  50. Vinciguerra, L.; Lanza, G.; Puglisi, V.; Pennisi, M.; Cantone, M.; Bramanti, A.; Pennisi, G.; Bella, R. Transcranial Doppler ultrasound in vascular cognitive impairment-no dementia. PLoS ONE 2019, 14, e0216162. [Google Scholar] [CrossRef] [Green Version]
  51. Abbott, N.J.; Patabendige, A.A.K.; Dolman, D.E.M.; Yusof, S.R.; Begley, D.J. Structure and function of the blood-brain barrier. Neurobiol. Dis. 2010, 37, 13–25. [Google Scholar] [CrossRef]
  52. Zlokovic, B.V. The Blood-Brain Barrier in Health and Chronic Neurodegenerative Disorders. Neuron 2008, 57, 178–201. [Google Scholar] [CrossRef] [Green Version]
  53. Cserr, H.F.; DePasquale, M.; Patlak, C.S. Regulation of brain water and electrolytes during acute hyperosmolality in rats. Am. J. Physiol. 1987, 253, F522–F529. [Google Scholar] [CrossRef] [PubMed]
  54. Fraser, P.A.; Dallas, A.D. Permeability of disrupted cerebral microvessels in the frog. J. Physiol. 1993, 461, 619–663. [Google Scholar] [CrossRef] [Green Version]
  55. Bridges, L.R.; Andoh, J.; Lawrence, A.; Khoong, C.H.; Poon, W.W.; Esiri, M.M.; Markus, H.S.; Hainsworth, A.H. Blood-Brain Barrier Dysfunction and Cerebral Small Vessel Disease (Arteriolosclerosis) in Brains of Older People. J. Neuropathol. Exp. Neurol. 2014, 73, 1026–1033. [Google Scholar] [CrossRef]
  56. Seo, J.H.; Miyamoto, N.; Hayakawa, K.; Pham, L.-D.D.; Maki, T.; Ayata, C.; Kim, K.-W.; Lo, E.H.; Arai, K. Oligodendrocyte precursors induce early blood-brain barrier opening after white matter injury. J. Clin. Investig. 2013, 123, 782–786. [Google Scholar] [CrossRef] [Green Version]
  57. Wardlaw, J.M.; Sandercock, P.A.G.; Dennis, M.S.; Starr, J. Ithe s breakdown of the blood-brain barrier responsible for lacunar stroke, leukoaraiosis, and dementia? Stroke 2003, 34, 806–812. [Google Scholar] [CrossRef]
  58. Wardlaw, J.M.; Doubal, F.N.; Valdes-Hernandez, M.; Wang, X.; Chappell, F.M.; Shuler, K.; Armitage, P.A.; Carpenter, T.C.; Dennis, M.S. Blood–Brain Barrier Permeability and Long-Term Clinical and Imaging Outcomes in Cerebral Small Vessel Disease. Stroke 2013, 44, 525–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Erdo, F.; Denes, L.; de Lange, E. Age-associated physiological and pathological changes at the blood-brain barrier: A review. J. Cereb. Blood Flow Metab. 2017, 37, 4–24. [Google Scholar] [CrossRef] [Green Version]
  60. Farrall, A.J.; Wardlaw, J.M. Blood-brain barrier: Aging and microvascular disease–systematic review and meta-analysis. Neurobiol. Aging 2009, 30, 337–352. [Google Scholar] [CrossRef] [PubMed]
  61. Li, Y.; Li, M.; Zhang, X.; Shi, Q.; Yang, S.; Fan, H.; Qin, W.; Yang, L.; Yuan, J.; Jiang, T.; et al. Higher blood-brain barrier permeability is associated with ha igher white matter hyperintensities burden. J. Neurol. 2017, 264, 1474–1481. [Google Scholar] [CrossRef] [PubMed]
  62. Li, Y.; Li, M.; Zuo, L.; Shi, Q.; Qin, W.; Yang, L.; Jiang, T.; Hu, W. Compromised blood-brain barrier integrity is associated with the total magnetic resonance imaging burden of cerebral small vessel disease. Front. Neurol. 2018, 9, 221. [Google Scholar] [CrossRef] [PubMed]
  63. Stringer, M.S.; Heye, A.K.; Armitage, P.A.; Chappell, F.; Hernández, M.D.C.V.; Makin, S.D.J.; Sakka, E.; Thrippleton, M.J.; Wardlaw, J.M. Tracer kinetic assessment of blood-brain barrier leakage and blood volume in cerebral small vessel disease: Associations with disease burden and vascular risk factors. NeuroImage 2021, 32, 102883. [Google Scholar] [CrossRef]
  64. Thrippleton, M.J.; Backes, W.H.; Sourbron, S.; Ingrisch, M.; Osch, M.J.P.; Dichgans, M.; Fazekas, F.; Ropele, S.; Frayne, R.; Oostenbrugge, R.J.; et al. Quantifying blood-brain barrier leakage in small vessel disease: Review and consensus recommendations. Alzheimer’s Dement. 2019, 15, 840–858. [Google Scholar] [CrossRef] [PubMed]
  65. Manning, C.; Stringer, M.; Dickie, B.; Clancy, U.; Hernandez, M.C.V.; Wiseman, S.J.; Garcia, D.J.; Sakka, E.; Backes, W.H.; Ingrisch, M.; et al. Sources of systematic error in DCE-MRI estimation of low-level blood-brain barrier leakage. Magn. Reason. Med. 2021, 86, 1888–1903. [Google Scholar] [CrossRef]
  66. Stewart, C.R.; Stringer, M.S.; Shi, Y.; Thrippleton, M.J.; Wardlaw, J.M. Associations Between White Matter Hyperintensity Burden, Cerebral Blood Flow and Transit Time in Small Vessel Disease: An Updated Meta-Analysis. Front. Neurol. 2021, 12, 647848. [Google Scholar] [CrossRef] [PubMed]
  67. Heye, A.K.; Thrippleton, M.J.; Armitage, P.A.; Hernandez, M.D.C.V.; Makin, S.D.; Glatz, A.; Sakka, E.; Wardlaw, J.M. Tracer kinetic modelling for DCE-MRI quantification of subtle blood-brain barrier permeability. Neuroimage 2016, 125, 446–455. [Google Scholar] [CrossRef] [Green Version]
  68. Mäe, M.A.; He, L.; Nordling, S.; Vazquez-Liebanas, E.; Nahar, K.; Jung, B.; Li, X.; Tan, B.C.; Chin Foo, J.; Cazenave-Gassiot, A.; et al. Single-Cell Analysis of Blood-Brain Barrier Response to Pericyte Loss. Circ. Res. 2021, 128, e46–e62. [Google Scholar] [CrossRef]
  69. Zhang, E.T.; Inman, C.B.; Weller, R.O. Interrelationships of the pia mater and the perivascular (Wirchov- Robin) spaces in the human cerebrum. J. Anat. 1990, 170, 111–123. [Google Scholar]
  70. Iadecola, C. The neurovascular Unit coming of age: A journey through neurovascular coupling in health and disease. Neuron 2017, 96, 17–42. [Google Scholar] [CrossRef] [Green Version]
  71. Hendrikx, D.; Smits, A.; Lavanga, M.; De Wel, O.; Thewissen, L.; Jansen, K.; Caicedo, A.; Van Huffe, S.; Naulaers, G. Measurement of Neurovascular Coupling in Neonates. Front. Physiol. 2019, 10, 65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Sweeney, M.D.; Sagare, A.P.; Zlokovic, B.V. Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat. Rev. Neurol. 2018, 14, 133–150. [Google Scholar] [CrossRef]
  73. Abbott, N.J.; Pizzo, M.E.; Preston, J.E.; Janigro, D.; Thorne, R.G. The role of brain barriers in fluid movement in the CNS: Is there a ‘glymphatic’ system? Acta Neuropathol. 2018, 135, 387–407. [Google Scholar] [CrossRef] [Green Version]
  74. Huijts, M.; Duits, A.; Staals, J.; Kroon, A.A.; De Leeuw, P.W.; Van Oostenbrugge, R.J. Basal ganglia enlarged perivascular spaces are linked to cognitive function in patients with cerebral small vessel disease. Curr. Neurovasc. Res. 2014, 11, 136–141. [Google Scholar] [CrossRef] [PubMed]
  75. Dalkara, T.; Alarcon-Martinez, L. Cerebral micro-vascular signaling in health and disease. Brain Res. 2015, 1623, 3–17. [Google Scholar] [CrossRef] [PubMed]
  76. Wardlaw, J.M.; Benveniste, H.; Nedergaard, M.; Zlokovic, B.V.; Mestre, H.; Lee, H.; Doubal, F.N.; Brown, R.; Ramirez, J.; MacIntosh, B.J.; et al. Perivascular spaces in the brain: Anatomy, physiology and pathology. Nat. Rev. Neurol. 2020, 16, 137–153. [Google Scholar] [CrossRef]
  77. Zhang, W.; Zhou, Y.; Wang, J.; Gong, X.; Chen, Z.; Zhang, X.; Cai, J.; Chen, S.; Fang, L.; Sun, J.; et al. Glymphatic clearance function in patients with cerebral small vessel disease. Neuroimage 2021, 238, 118257. [Google Scholar] [CrossRef]
  78. Benveniste, H.; Nedergaard, M. Cerebral small vessel disease: A glymphopathy? Curr. Opin. Neurobiol. 2022, 72, 15–21. [Google Scholar] [CrossRef]
  79. Jiménez-Balado, J.; Riba-Llena, I.; Garde, E.; Valor, M.; Gutiérrez, B.; Pujadas, F.; Delgado, P. Prevalence of hippocampal enlarged perivascular spaces in a sample of patients with hypertension and their relation with vascular risk factors and cognitive function. J. Neurol. Neurosurg. Psychiatry 2018, 89, 651–656. [Google Scholar] [CrossRef]
  80. Giannakopoulos, P.; Gold, G.; Kowaru, E.; von Gunten, A.; Imhof, A.; Bouras, C. Assessing the cognitive impact of Alzheimer disease pathology and vascular burden in the aging brain: The Geneva experience. Acta Neuropathol. 2007, 113, 1–12. [Google Scholar] [CrossRef]
  81. Van der Veen, P.H.; Muller, M.; Vinken, K.L.; Hendrikse, J.; Mali, W.P.; van der Graaf, Y.; Geerlings, M.I.; SMART Study Group. Longitudinal relationship between cerebral small vessel disease and cerebral blood flow. The second manifestations of arterial disease-magnetic resonance study. Stroke 2015, 46, 1233–1238. [Google Scholar] [CrossRef] [Green Version]
  82. Cuadrado-Godia, E.; Dwivedi, P.; Sharma, S.; Santiago, A.O.; Roquer Gonzalez, J.; Balcells, M.; Laird, J.; Turk, M.; Suri, H.S.; Nicolaides, A.; et al. Cerebral Small Vessel Disease: A Review Focusing on Pathophysiology, Biomarkers, and Machine Learning Strategies. J. Stroke 2018, 20, 302–320. [Google Scholar] [CrossRef]
  83. Zhang, C.E.; Wong, S.M.; van de Haar, H.J.; Staals, J.; Jansen, J.F.; Jeukens, C.R.; Hofman, P.A.; van Oostenbrugge, R.J.; Backes, W.H. Blood-brain barrier leakage is more widespread in patients with cerebral small vessel disease. Neurology 2017, 88, 426–432. [Google Scholar] [CrossRef] [PubMed]
  84. Huisa, B.N.; Caprihan, A.; Thompson, J.; Prestopnik, J.; Qualls, C.R.; Rosenberg, G.A. Long-term blood-brain barrier permeability changes in Binswanger disease. Stroke 2015, 46, 2413–2418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Wardlaw, J.M.; Makin, S.J.; Hernández, M.C.V.; Armitage, P.A.; Heye, A.K.; Chappell, F.M.; Muñoz-Maniega, S.; Sakka, E.; Shuler, K.; Dennis, M.S.; et al. Blood-brain barrier failure as a core mechanism in cerebral small vessel disease and dementia: Evidence from a cohort study. Alzheimer’s Dement. 2017, 13, 634–643. [Google Scholar] [CrossRef] [Green Version]
  86. Ihara, M.; Yamamoto, Y. Emerging evidence for pathogenesis of sporadic cerebral small vessel disease. Stroke 2016, 47, 554–560. [Google Scholar] [CrossRef] [Green Version]
  87. Rajani, R.M.; Williams, A. Endothelial cell-oligodendrocyte interactions in small vessel disease and aging. Clin. Sci. 2017, 131, 369–379. [Google Scholar] [CrossRef] [Green Version]
  88. Furukawa, S.; Sameshima, H.; Yang, L.; Hariskuma, M.; Ikenoue, T. Regional differences of microglial accumulation within 72 hours of hypoxia-ischemia and the effect of acetylcholine receptor agonist on brain damage and microglial activation in newborn rats. Brain Res. 2014, 1562, 52–58. [Google Scholar] [CrossRef]
  89. Petito, C.K. Transformation of postisichemic perineuronal glial cells. J. Cereb. Blood Flow Metabol. 1986, 6, 616–624. [Google Scholar] [CrossRef] [Green Version]
  90. Petito, C.K.; Olarte, J.P.; Roberts, B.; Nowak, T.S.; Pulsinelli, W.A. Selective glial vulnerability following transient global ischemia in rat brain. J. Neuropathol. Exp. Neurol. 1998, 57, 231–238. [Google Scholar] [CrossRef]
  91. Masuda, T.; Croom, D.; Hida, H.; Kirov, S.A. Capillary blood flow around microglial somata determines dynamics of microglial processes in ischemic conditions. Glia 2011, 59, 1744–1753. [Google Scholar] [CrossRef] [Green Version]
  92. Ju, F.; Ran, Y.; Zhu, L.; Cheng, X.; Gao, H.; Xi, X.; Yang, Z.; Zhang, S. Increased BBB Permeability Enhances Activation of Microglia and Exacerbates Loss of Dendritic Spines after Transient Global Cerebral Ischemia. Front. Cell Neurosci. 2018, 12, 236. [Google Scholar] [CrossRef]
  93. Zhang, S. Microglial activation after ischaemic stroke. Stroke Vasc. Neurol. 2019, 4, 71–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Iadecola, C. The pathobiology of vascular dementia. Neuron 2013, 80, 844–866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Filous, A.S.; Silver, J. Targeting astrocytes in CNS injury and disease: A translational research approach. Prog. Neurobiol. 2016, 144, 173–187. [Google Scholar] [CrossRef] [Green Version]
  96. Forsberg, K.M.E.; Zhang, Y.; Reiners, J.; Ander, M.; Niedermayer, A.; Fang, L.; Neugebauer, H.; Kassubek, J.; Katona, I.; Weis, J.; et al. Endothelial damage, vascular bagging and remodeling of the microvascular bed in human microangiopathy with deep white matter lesions. Acta Neuropathol. Commun. 2018, 6, 128. [Google Scholar] [CrossRef] [PubMed]
  97. Szalay, G.; Martinecz, B.; Lénárt, N.; Környei, Z.; Orsolits, B.; Judák, L.; Császár, E.; Fekete, R.; West, B.L.; Katona, G.; et al. Microglia protect against brain injury and their selective elimination dysregulates neuronal network activity after stroke. Nat. Commun. 2016, 7, 11499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Zhao, S.-C.; Ma, L.-S.; Chu, Z.-H.; Xu, H.; Wu, W.-Q.; Liu, F. Regulation of microglial activation in stroke. Acta Pharmacol. Sin. 2017, 38, 445–458. [Google Scholar] [CrossRef] [PubMed]
  99. Morrison, H.W.; Filosa, J.A. A quantitative spatiotemporal analysis of microglia morphology during ischemic stroke and reperfusion. J. Neuroinflamm. 2013, 10, 4. [Google Scholar] [CrossRef] [Green Version]
  100. Perego, C.; Fumagalli, S.; De Simoni, M.G. Temporal pattern of expression and colocalization of microglia/macrophage phenotype markers following brain ischemic injury in mice. J. Neuroinflamm. 2011, 8, 174. [Google Scholar] [CrossRef] [Green Version]
  101. Farkas, E.; Donka, G.; de Vous, R.A.I.; Mihaly, A.; Bari, F.; Luiten, P.G.M. Experimental cerebral hypoperfusion induces white matter injury and microglial activation in the rat brain. Acta Neuropathol. 2004, 108, 57–64. [Google Scholar] [CrossRef]
  102. Paolini Paoletti, F.; Simoni, S.; Parnetti, L.; Gaetani, L. The Contribution of Small Vessel Disease to Neurodegeneration: Focus on Alzheimer’s Disease, Parkinson’s Disease and Multiple Sclerosis. Int. J. Mol. Sci. 2021, 22, 4958. [Google Scholar] [CrossRef]
  103. Fredman, P.; Wallin, A.; Blennow, K.; Davidsson, P.; Gottfries, C.; Svennerholm, L. Sulfatide as a biochemical marker in cerebrospinal fluid of patients with vascular dementia. Acta Neurol. Scand. 1992, 85, 103–106. [Google Scholar] [CrossRef] [PubMed]
  104. Tullberg, M.; Månsson, J.E.; Fredman, P.; Lekman, A.; Blennow, K.; Ekman, R.; Rosengren, L.E.; Tisell, M.; Wikkelso, C. CSF sulfatide distinguishes between normal pressure hydrocephalus and subcortical arteriosclerotic encephalopathy. J. Neurol. Neurosurg. Psychiatry 2000, 69, 74–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Thibert, K.A.; Raymond, G.V.; Nascene, D.R.; Miller, W.P.; Tolar, J.; Orchard, P.J.; Lund, T.C. Cerebrospinal fluid matrix metalloproteinases are elevated in cerebral adrenoleukodystrophy and correlate with MRI severity and neurologic dysfunction. PLoS ONE 2012, 7, e50430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Jonsson, M.; Zetterberg, H.; Van Straaten, E.; Lind, K.; Syversen, S.; Edman, Å.; Blennow, K.; Rosengren, L.; Pantoni, L.; Inzitari, D.; et al. Cerebrospinal fluid biomarkers of white matter lesions—Cross-sectional results from the LADIS study. Eur. J. Neurol. 2010, 17, 377–382. [Google Scholar] [CrossRef] [PubMed]
  107. Bjerke, M.; Andreasson, U.; Rolstad, S.; Nordlund, A.; Lind, K.; Zetterberg, H.; Edman, Å.; Blennow, K.; Wallin, A. Subcortical vascular dementia biomarker pattern in mild cognitive impairment. Dement. Geriatr. Cogn. Disord. 2009, 28, 348–356. [Google Scholar] [CrossRef]
  108. Öhrfelt, A.; Andreasson, U.; Simon, A.; Zetterberg, H.; Edman, Å.; Potter, W.; Holder, D.; Devanarayan, V.; Seeburger, J.; Smith, A.D.; et al. Screening for New Biomarkers for Subcortical Vascular Dementia and Alzheimer’s Disease. Dement. Geriatr. Cogn. Dis. Extra 2011, 1, 31–42. [Google Scholar] [CrossRef]
  109. Peters, A.; Sethares, C. Age-related changes in the morphology of cerebral capillaries do not correlate with cognitive decline. J. Comp. Neurol. 2012, 520, 1339–1347. [Google Scholar] [CrossRef]
  110. Iejima, D.; Itabashi, T.; Kawamura, Y.; Noda, T.; Yuasa, S.; Fukuda, K.; Oka, C.; Iwata, T. HTRA1 (high temperature requirement A serine peptidase 1) gene is transcriptionally regulated by insertion/deletion nucleotides located at the 3′ end of the ARMS2 (age related maculopathy susceptibility 2) gene in patients with age-related macular degeneration. J. Biol. Chem. 2015, 290, 2784–2797. [Google Scholar]
  111. Zlokovic, B.V. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat. Rev. Neurosci. 2011, 12, 723–728. [Google Scholar] [CrossRef]
  112. Cai, W.; Zhang, K.; Li, P.; Zhu, L.; Xu, J.; Yang, B.; Hu, X.; Lu, Z.; Chen, J. Dysfunction of the neurovascular unit in ischemic stroke and neurodegenerative diseases: An aging effect. Ageing Res. Rev. 2017, 34, 77–87. [Google Scholar] [CrossRef] [Green Version]
  113. Prisby, R.D.; Ramsey, M.W.; Behnke, B.J.; Dominguez, J.M.; Donato, A.J.; Allen, M.R.; Delp, M.D. Aging reduces skeletal blood flow endothelium dependent vasodilation, and NO bioavailability in Rats. J. Bone Miner. Res. 2007, 22, 1280–1288. [Google Scholar] [CrossRef] [PubMed]
  114. Nicholson, W.T.; Vaa, B.; Hesse, C.; Eisenach, J.H.; Joyner, M.J. Aging is associated with reduced prostacyclin-mediated dilation in the human forearm. Hypertension 2009, 53, 973–978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Van Dijk, E.J.; Prins, N.D.; Vermeer, S.E.; Vrooman, H.A.; Hofman, A.; Koudstaal, P.J.; Breteler, M.M.B. C-reactive protein and cerebral small-vessel disease: The Rotterdam Scan Study. Circulation 2005, 112, 900–905. [Google Scholar] [CrossRef] [Green Version]
  116. Long, D.A.; Newaz, M.A.; Prabahakar, S.S.; Price, K.L.; Truong, L.; Feng, L.; Mu Oyekan, A.O.; Johnson, R.J. Loss of nitric oxide and endothelial-derived hyperpolarizing factor-mediated responses in ageing. Kidney Int. 2005, 68, 2154–2163. [Google Scholar] [CrossRef] [Green Version]
  117. Van der Loo, B.; Labugger, R.; Skepper, J.N.B.; Achschmid, M.; Kilo, J.; Powell, J.M.; Palacios-Callendere, M.; Erusalimsky, J.D.; Quaschning, T.; Malinski, T. Enhanced peroxynitrite formation is associated with vascular ageing. J. Exp. Med. 2000, 18, 1731–1744. [Google Scholar] [CrossRef] [Green Version]
  118. Puca, A.A.; Carrizzo, A.; Ferrario, A.; Villa, F.; Vecchione, C. Endothelial nitric oxide synthase, vascular integrity and human exceptional longevity. Immun. Ageing 2012, 9, 26. [Google Scholar] [CrossRef] [Green Version]
  119. Flentje, A.; Kalsi, R.; Monahan, T.S. Small GTPases and Their Role in Vascular Disease. Int. J. Mol. Sci. 2019, 20, 917. [Google Scholar] [CrossRef] [Green Version]
  120. Hartmann, S.; Ridley, A.J.; Lutz, S. The Function of Rho-Associated Kinases ROCK1 and ROCK2 in the Pathogenesis of Cardiovascular Disease. Front. Pharmacol. 2015, 6, 276. [Google Scholar] [CrossRef]
  121. Szulcek, R.; Beckers, C.M.; Hodzic, J.; de Wit, J.; Chen, Z.; Grob, T.; Musters, R.J.; Minshall, R.D.; van Hinsbergh, V.W.; van Nieuw Amerongen, G.P. Localized RhoA GTPase activity regulates dynamics of endothelial monolayer integrity. Cardiovasc. Res. 2013, 99, 471–482. [Google Scholar] [CrossRef] [Green Version]
  122. Van Nieuw Amerongen, G.P.; Beckers, C.M.; Achekar, I.D.; Zeeman, S.; Musters, R.J.; van Hinsbergh, V.W. Involvement of Rho kinase in endothelial barrier maintenance. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 2332–2339. [Google Scholar] [CrossRef] [Green Version]
  123. Wang, J.; Liu, H.; Chen, B.; Li, Q.; Huang, X.; Wang, L.; Guo, X.; Huang, Q. RhoA/ROCK-dependent moesin phosphorylation regulates AGE-induced endothelial cellular response. Cardiovasc. Diabetol. 2012, 11, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Sun, H.; Breslin, J.W.; Zhu, J.; Yuan, S.Y.; Wu, M.H. Rho and ROCK signaling in VEGF-induced microvascular endothelial hyperpermeability. Microcirculation 2006, 13, 237–247. [Google Scholar] [CrossRef] [PubMed]
  125. Simpson, J.E.; Fernando, M.S.; Clark, L.; Ince, P.G.; Matthews, F.; Forster, G.; O’Brien, J.T.; Barber, R.; Kalaria, R.N.; Brayne, C.; et al. White matter lesions in an unselected cohort of the elderly: Astrocytic, microglial and oligodendrocyte precursor cell responses. Neuropathol. Appl. Neurobiol. 2007, 33, 410–419. [Google Scholar] [CrossRef] [PubMed]
  126. Sies, H.; Jones, D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef]
  127. Pantoni, L.; Inzitari, D.; Pracucci, G.; Lolli, F.; Giordano, G.; Bracco, L.; Amaducci, L. Cerebrospinal fluid proteins in patients with leucoaraiosis: Possible abnormalities in blood-brain barrier function. J. Neurol. Sci. 1993, 115, 125–131. [Google Scholar] [CrossRef]
  128. Musaeus, C.S.; Gleerup, H.S.; Høgh, P.; Waldemar, G.; Hasselbalch, S.G.; Simonsen, A.H. Cerebrospinal Fluid/Plasma Albumin Ratio as a Biomarker for Blood-Brain Barrier Impairment Across Neurodegenerative Dementias. J. Alzheimers Dis. 2020, 75, 429–436. [Google Scholar] [CrossRef]
  129. Georgakis, M.K.; Chatzopoulou, D.; Tsivgoulis, G.; Petridou, E.T. Albuminuria and cerebral small vessel disease: A systematic review and meta-analysis. J. Am. Geriatr. Soc. 2018, 66, 509–517. [Google Scholar] [CrossRef]
  130. Wada, M.; Takahashi, Y.; Iseki, C.; Kawanami, T.; Daimon, M.; Kato, T. Plasma fibrinogen, global cognitive function, and cerebral small vessel disease: Results of a cross-sectional study in community-dwelling Japanese elderly. Intern. Med. 2011, 50, 999–1007. [Google Scholar] [CrossRef] [Green Version]
  131. Kulikauskas, M.R.; Shaka, X.; Bautch, V.L. The versatility and paradox of BMP signaling in endothelial cell behaviors and blood vessel function. Cell. Mol. Life Sci. 2022, 79, 77. [Google Scholar] [CrossRef]
  132. Knottnerus, I.L.; Govers-Riemslag, J.W.; Hamulyak, K.; Rouhl, R.P.; Staals, J.; Spronk, H.M. Endothelial activation in lacunar stroke subtypes. Stroke 2010, 41, 1617–1622. [Google Scholar] [CrossRef] [Green Version]
  133. Knottnerus, I.L.; Cate, H.; Lodder, J.; Kessels, F.; van Oostenbrugge, R.J. Endothelial dysfunction in lacunar stroke: A systematic review. Cerebrovasc. Dis. 2009, 27, 519–526. [Google Scholar] [CrossRef] [PubMed]
  134. Stevenson, S.F.; Doubal, F.N.; Shuler, K.; Wardlaw, J.M. A systematic review of dynamic cerebral andperipheral endothelial function in lacunar stroke versus controls. Stroke 2010, 41, e434–e442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Markus, H.S.; Hunt, B.; Palmer, K.; Enzinger, C.; Schmidt, H.; Schmidt, R. Markers of endothelial and hemostatic activation and progression of cerebral white matter hyperintensities: Longitudinal results of the Austrian Stroke Prevention Study. Stroke 2005, 36, 1410–1414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Fernando, M.S.; Simpson, J.E.; Matthews, F.; Brayne, C.; Lewis, C.E.; Barber, R.; Kalaria, R.N.; Forster, G.; Esteves, F.; Wharton, S.B.; et al. White matter lesions in an unselected cohort of the elderly: Molecular pathology suggests origin from chronic hypoperfusion injury. Stroke 2006, 37, 1391–1398. [Google Scholar] [CrossRef] [Green Version]
  137. Egea, J.; Fabregat, I.; Frapart, Y.M.; Ghezzi, P.; Görlach, A.; Kietzmann, T.; Kubaichuk, K.; Knaus, U.G.; Lopez, M.G.; Olaso-Gonzalez, G.; et al. European contribution to the study of ROS: A summary of the findings and prospects for the future from the COST action BM1203 (EU-ROS). Redox Biol. 2017, 13, 94–162, Erratum in Redox Biol. 2018, 14, 694–696. [Google Scholar] [CrossRef] [Green Version]
  138. Dikalov, S.I.; Dikalova, A.E.; Bikineyeva, A.T.; Schmidt, H.H.; Harrison, D.G.; Griendling, K.K. Distinct roles of Nox1 and Nox4 in basal and angiotensin II-stimulated superoxide and hydrogen peroxide production. Free Radic. Biol. Med. 2008, 45, 1340–1351. [Google Scholar] [CrossRef] [Green Version]
  139. Zhang, X.; Wu, B.; Nie, K.; Jia, Y.; Yu, J. Effects of acupuncture on declined cerebral blood flow, impaired mitochondrial respiratory function and oxidative stress in multi-infarct dementia rats. Neurochem. Int. 2014, 65, 23–29. [Google Scholar] [CrossRef]
  140. Huang, J.L.; Fu, S.T.; Jiang, Y.Y.; Cao, Y.B.; Guo, M.L.; Wang, Y.; Xu, Z. Protective effects of Nicotiflorin on reducing memory dysfunction, energy metabolism failure, and oxidative stress in multi-infarct dementia model rats. Pharmacol. Biochem. Behav. 2007, 86, 741–748. [Google Scholar] [CrossRef]
  141. Takac, I.; Schröder, K.; Brandes, R.P. The Nox family of NADPH oxidases: Friend or foe of the vascular system? Curr. Hypertens. Rep. 2012, 14, 70–78. [Google Scholar] [CrossRef]
  142. Miller, A.A.; Drummond, G.R.; DeSilva, T.M.; Mast, A.E.; Hickey, H.; Williams, J.P.; Broughton, B.R.; Sobey, C.G. NADPH oxidase activity is higher in cerebral versus systemic arteries of four animal species: Role of Nox2. Am. J. Physiol. Heart Circ. Physiol. 2009, 296, H220–H225. [Google Scholar] [CrossRef]
  143. Liu, H.; Zhang, J. Cerebral hypoperfusion and cognitive impairment: The pathogenic role of vascular oxidative stress. Int. J. Neurosci. 2012, 122, 494–499. [Google Scholar] [CrossRef] [PubMed]
  144. Paravicini, T.M.; Miller, A.A.; Drummond, G.R.; Sobey, C.G. Flow-induced cerebral vasodilatation in vivo involves activation of phosphatidylinositol-3kinase, NADPH-oxidase, and nitric oxide synthase. J. Cereb. Blood Flow Metab. 2006, 26, 836–845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. De Silva, T.M.; Brait, V.H.; Drummond, G.R.; Sobey, C.G.; Miller, A.A. Nox2 oxidase activity accounts for the oxidative stress and vasomotor dysfunction in mouse cerebral arteries following ischemic stroke. PLoS ONE 2011, 6, e28393. [Google Scholar] [CrossRef] [PubMed]
  146. Lin, M.T.; Beal, M.F. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature 2006, 443, 787–795. [Google Scholar] [CrossRef]
  147. Raz, N. The aging brain: Structural changes and their implications for cognitive aging. In New Frontiers in Cognitive Aging; Dixon, R., Bäckman, L., Nilsson, L., Eds.; Oxford University Press: Telangana, India, 2004; pp. 115–134. [Google Scholar]
  148. Sun, N.; Youle, R.J.; Finkel, T. The mitochondrial basis of aging. Mol. Cell 2016, 61, 654–666. [Google Scholar] [CrossRef] [Green Version]
  149. Valiente-Pallejà, A.; Tortajada, J.; Bulduk, B.K.; Vilella, E.; Garrabou, G.; Muntané, G.; Martorell, L. Comprehensive summary of mitochondrial DNA alterations in the postmortem human brain: A systematic review. EBioMedicine 2022, 76, 103815. [Google Scholar] [CrossRef]
  150. Corral-Debrinski, M.; Horton, T.; Lott, M.T.; Shoffner, J.M.; Beal, M.F.; Wallace, D.C. Mitochondrial DNA deletions in human brain: Regional variability and increase with advanced age. Nat. Genet. 1992, 2, 324–329. [Google Scholar] [CrossRef]
  151. Taylor, S.D.; Ericson, N.G.; Burton, J.N.; Prolla, T.A.; Silber, J.R.; Shendure, J.; Bielas, J.H. Targeted enrichment and high-resolution digital profiling of mitochondrial DNA deletions in human brain. Aging Cell 2014, 13, 29–38. [Google Scholar] [CrossRef]
  152. Kennedy, S.R.; Salk, J.J.; Schmitt, M.W.; Loeb, L.A. Ultra-sensitive sequencing reveals an age-related increase in somatic mitochondrial mutations that are inconsistent with oxidative damage. PLoS Genet. 2013, 9, 1003794. [Google Scholar] [CrossRef] [Green Version]
  153. Diaz, F.; Bayona-Bafaluy, M.P.; Rana, M.; Mora, M.; Hao, H.; Moraes, C.T. Human mitochondrial DNA with large deletions repopulates organelles faster than full-length genomes under relaxed copy number control. Nucleic Acids Res. 2002, 30, 4626–4633. [Google Scholar] [CrossRef]
  154. Guo, L.; Tian, J.; Du, H. Mitochondrial dysfunction and synaptic transmission failure in Alzheimer’s disease. J. Alzheimers Dis. 2017, 57, 1071–1086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Ballif, B.C.; Theisen, A.; Coppinger, J.; Gowans, G.C.; Hersh, J.H.; Madan-Khetarpal, S.; Schmidt, K.R.; Tervo, R.; Escobar, L.F.; Friedrich, C.A.; et al. Expanding the clinical phenotype of the 3q29 microdeletion syndrome and characterization of the reciprocal microduplication. Mol. Cytogenet. 2008, 1, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Lanza, G.; Cantone, M.; Musso, S.; Borgione, E.; Scuderi, C.; Ferri, R. Early-onset subcortical ischemic vascular dementia in an adult with mtDNA mutation 3316G>A. J. Neurol. 2018, 265, 968–969. [Google Scholar] [CrossRef] [PubMed]
  157. Campbell, G.R.; Ziabreva, I.; Reeve, A.K.; Krishnan, K.J.; Reynolds, R.; Howell, O.; Lassmann, H.; Turnbull, D.M.; Mahad, D.J. Mitochondrial DNA deletions and neurodegeneration in multiple sclerosis. Ann. Neurol. 2011, 69, 481–492. [Google Scholar] [CrossRef] [Green Version]
  158. Chinnery, P.F. Mitochondrial disorders overview. In GeneReviews [Internet]; Margaret, P.A., Ardinger, H.H., Pagon, R.A., Wallace, S.E., Bean, L.J.H., Karen, S.A.A., Eds.; University of Washington, Seattle: Seattle, WA, USA, 2020; p. 1993. [Google Scholar]
  159. Basel, D. Mitochondrial DNA Depletion Syndromes. Clin. Perinatol. 2020, 47, 123–141. [Google Scholar] [CrossRef]
  160. Coskun, P.E.; Wyrembak, J.; Derbereva, O.; Melkonian, G.; Doran, E.; Lott, I.T.; Head, E.; Cotman, C.W.; Wallace, D.C. Systemic mitochondrial dysfunction and the etiology of Alzheimer’s disease and down syndrome dementia. J. Alzheimers Dis. 2010, 20, 293–310. [Google Scholar] [CrossRef] [Green Version]
  161. Roca-Bayerri, C.; Robertson, F.; Pyle, A.; Hudson, G.; Payne, B.A.I. Mitochondrial DNA damage and brain aging in human immunodeficiency virus. Clin. Infect. Dis. 2021, 73, e466–e473. [Google Scholar] [CrossRef]
  162. Touyz, R.M.; Briones, A.M. Reactive oxygen species and vascular biology: Implications in human hypertension. Hypertens. Res. 2011, 34, 5–14. [Google Scholar] [CrossRef] [Green Version]
  163. Mayhan, W.G.; Arrick, D.M.; Sharpe, G.M.; Sun, H. Age-related alterations in reactivity of cerebral arterioles: Role of oxidative stress. Microcirculation 2008, 15, 225–236. [Google Scholar] [CrossRef]
  164. Dong, Y.F.; Kataoka, K.; Toyama, K.; Sueta, D.; Koibuchi, N.; Yamamoto, E.; Yata, K.; Tomimoto, H.; Ogawa, H.; Kim-Mitsuyama, S. Attenuation of brain damage and cognitive impairment by direct renin inhibition in mice with chronic cerebral hypoperfusion. Hypertension 2011, 58, 635–642. [Google Scholar] [CrossRef] [Green Version]
  165. Santhanam, A.V.; d’Uscio, L.V.; Katusic, Z.S. Erythropoietin increases bioavailability of tetrahydrobiopterin and protects cerebral microvasculature against oxidative stress induced by eNOS uncoupling. J. Neurochem. 2014, 131, 521–529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Xie, H.; Ray, P.E.; Short, B.L. NF-kappa B activation plays a role in superoxide-mediated cerebral dysfunction after hypoxia/reoxygenation. Stroke 2005, 36, 1047–1052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Aghajanian, A.; Wittchen, E.S.; Campbell, S.L.; Burridge, K. Direct activation of RhoA by reactive oxygen species requires a redox-sensitive motif. PLoS ONE 2009, 4, e8045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Faraco, G.; Moraga, A.; Moore, J.; Anrather, J.; Pickel, V.M.; Iadecola, C. Circulating endothelin-1 alters critical mechanisms regulating cerebral microcirculation. Hypertension 2013, 62, 759–766. [Google Scholar] [CrossRef]
  169. Bochkov, V.N.; Philippova, M.; Oskolkova, O.; Kadl, A.; Furnkranz, A.; Karabeg, E.; Afonyushkin, T.; Gruber, F.; Breuss, J.; Minchenko, A.; et al. Oxidized phospholipids stimulate angiogenesis via autocrine mechanisms, implicating a novel role for lipid oxidation in the evolution of atherosclerotic lesions. Circ. Res. 2006, 99, 900–908. [Google Scholar] [CrossRef]
  170. Tai, L.M.; Thomas, R.; Marottoli, F.M.; Koster, K.P.; Kanekiyo, T.; Morris, A.W.; Bu, G. The role of APOE in cerebrovascular dysfunction. Acta Neuropathol. 2016, 131, 709–723. [Google Scholar] [CrossRef] [Green Version]
  171. Han, B.H.; Zhou, M.L.; Johnson, A.W.; Singh, I.; Liao, F.; Vellimana, A.K.; Nelson, J.W.; Milner, E.; Cirrito, J.R.; Basak, J.; et al. Contribution of reactive oxygen species to cerebral amyloid angiopathy, vasomotor dysfunction, and micro-hemorrhage in aged Tg2576 mice. Proc. Natl. Acad. Sci. USA 2015, 112, E881–E890. [Google Scholar] [CrossRef] [Green Version]
  172. Grochowski, C.; Litak, J.; Kamieniak, P.; Maciejewski, R. Oxidative stress in cerebral small vessel disease. Role of reactive species. Free Radic. Res. 2018, 52, 1–13. [Google Scholar] [CrossRef]
  173. Han, S.; Wu, H.; Li, W.; Gao, P. Protective effects of genistein in homocysteine-induced endothelial cell inflammatory injury. Mol. Cell. Biochem. 2015, 403, 43–49. [Google Scholar] [CrossRef]
  174. Fleszar, M.G.; Wiśniewski, J.; Zboch, M.; Diakowska, D.; Gamian, A.; Krzystek-Korpacka, M. Targeted metabolomic analysis of nitric oxide/L-arginine pathway metabolites in dementia: Association with pathology, severity, and structural brain changes. Sci. Rep. 2019, 9, 1376. [Google Scholar] [CrossRef]
  175. Gao, Q.; Fan, Y.; Mu, L.-Y.; Ma, L.; Song, Z.-Q.; Zhang, Y.-N. S100B and ADMA in cerebral small vessel disease and cognitive dysfunction. J. Neurol. Sci. 2015, 354, 27–32. [Google Scholar] [CrossRef] [PubMed]
  176. Vinciguerra, L.; Lanza, G.; Puglisi, V.; Fisicaro, F.; Pennisi, M.; Bella, R.; Cantone, M. Update on the Neurobiology of Vascular Cognitive Impairment: From Lab to Clinic. Int. J. Mol. Sci. 2020, 21, 2977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Li, J.-J.; Li, Q.; Du, H.-P. Homocysteine Triggers inflammatory responses in macrophages through inhibiting CSE-H2S signaling via DNA hypermethylation of CSE promoter. Int. J. Mol. Sci. 2015, 16, 12560–12577. [Google Scholar] [CrossRef] [PubMed]
  178. Moretti, R.; Giuffré, M.; Caruso, P.; Gazzin, S.; Tiribelli, C. Homocysteine in Neurology: A Possible Contributing Factor to Small Vessel Disease. Int. J. Mol. Sci. 2021, 22, 2051. [Google Scholar] [CrossRef]
  179. Ahmad, S.; Siddiqi, M.I. Insights from molecular modeling into the selective inhibition of cathepsin S by its inhibitor. J. Mol. Model. 2017, 23, 92. [Google Scholar] [CrossRef]
  180. Leng, Y.P.; Ma, Y.S.; Li, X.G.; Chen, R.F.; Zeng, P.Y.; Li, X.H.; Qiu, C.F.; Li, Y.P.; Zhang, Z.; Chen, A.F. l-Homocysteine-induced cathepsin V mediates the vascular endothelial inflammation in hyperhomocysteinaemia. Br. J. Pharmacol. 2018, 175, 1157–1172. [Google Scholar] [CrossRef]
  181. Moretti, R.; Dal Ben, M.; Gazzin, S.; Tiribelli, C. Homcysteine in neurology: From endothelium to neurodegeneration. Curr. Nutr. Food Sci. 2017, 13, 163–175. [Google Scholar] [CrossRef]
  182. Deng, J.; Lu, S.; Li, H. Homocysteine activates B cells via regulating PKM-2 dependent metabolic reprogramming. J. Immunol. 2017, 198, 170–183. [Google Scholar] [CrossRef] [Green Version]
  183. Kumar, A.; Palfrey, H.A.; Pathak, R.; Kadowitz, P.J.; Gettys, T.W.; Murthy, S.N. The metabolism and significance of homocysteine in nutrition and health. Nutr. Metab. 2017, 14, 78. [Google Scholar] [CrossRef] [Green Version]
  184. Dayal, S.; Wilson, K.M.; Leo, L.; Arning, E.; Bottiglieri, T.; Lentz, S.R. Enhanced susceptibility to arterial thrombosis in a murine model of hyperhomocysteinemia. Blood 2006, 108, 2237–2243. [Google Scholar] [CrossRef] [Green Version]
  185. Perla-Kajan, J.; Twardowski, T.; Jakubowski, H. Mechanisms of homocysteine toxicity in humans. Amino Acids 2007, 32, 561–572. [Google Scholar] [CrossRef] [PubMed]
  186. Li, T.; Chen, Y.; Li, J.; Yang, X.; Zhang, H.; Qin, X.; Hu, Y.; Mo, Z. Serum Homocysteine Concentration Is Significantly Associated with Inflammatory/Immune Factors. PLoS ONE 2015, 10, e0138099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Reddy, V.S.; Trinath, J.; Reddy, G.B. Implication of homocysteine in protein quality control processes. Biochimie 2019, 165, 19–31. [Google Scholar] [CrossRef] [PubMed]
  188. Heo, J.H.; Lucero, J.; Abumiya, T.; Koziol, J.A.; Copeland, B.R.; del Zoppo, G.J. Matrix metalloproteinases increase very early during experimental focal cerebral ischemia. J. Cereb. Blood Flow Metab. 1999, 19, 624–633. [Google Scholar] [CrossRef] [Green Version]
  189. Zhang, M.; Zhu, W.; Yun, W.; Wang, Q.; Cheng, M.; Zhang, Z.; Liu, X.; Zhou, X.; Xu, G. Correlation of matrix metalloproteinase-2 single nucleotide polymorphisms with the risk of small vessel disease (SVD). J. Neurol. Sci. 2015, 356, 61–64. [Google Scholar] [CrossRef]
  190. Yang, Y.; Estrada, E.Y.; Thompson, J.F.; Liu, W.; Rosenberg, G.A. Matrix metalloproteinase-mediated disruption of tight junction proteins in cerebral vessels is reversed by synthetic matrix metalloproteinase inhibitor in focal ischemia in rat. J. Cereb. Blood Flow Metab. 2007, 27, 697–709. [Google Scholar] [CrossRef]
  191. Stamenkovic, I. Extracellular matrix remodeling: The role of matrix metalloproteinases. J. Pathol. 2003, 200, 448–464. [Google Scholar] [CrossRef]
  192. English, W.R.; Suarez-Puente, X.S.; Freije, J.M.; Knauper, V.; Amour, A.; Merryweather, A.; López-Otín, C.; Murphy, G. Membrane type 4 matrix metalloproteinase (MMP17) has tumor necrosis factor-alpha convertase activity but does not activate pro-MMP2. J. Biol. Chem. 2000, 275, 14046–14055. [Google Scholar] [CrossRef] [Green Version]
  193. Kim, Y.S.; Choi, D.H.; Block, M.L.; Lorenzl, S.; Yang, L.; Kim, Y.J.; Sugama, S.; Cho, B.P.; Hwang, O.; Browne, S.E.; et al. A pivotal role of matrix metalloproteinase-3 activity in dopaminergic neuronal degeneration via microglial activation. FASEB J. 2007, 21, 179–187. [Google Scholar] [CrossRef] [Green Version]
  194. Woo, M.S.; Park, J.S.; Choi, I.Y.; Kim, W.K.; Kim, H.S. Inhibition of MMP-3 or -9 suppresses lipopolysaccharide-induced expression of proinflammatory cytokines and iNOS in microglia. J. Neurochem. 2008, 106, 770–780. [Google Scholar] [CrossRef]
  195. Powell, W.C.; Fingleton, B.; Wilson, C.L.; Boothby, M.; Matrisian, L.M. The metalloproteinase matrilysin proteolytically generates active soluble Fas ligand and potentiates epithelial cell apoptosis. Curr. Biol. 1999, 9, 1441–1447. [Google Scholar] [CrossRef] [Green Version]
  196. Gu, Y.; Zheng, G.; Xu, M.; Li, Y.; Chen, X.; Zhu, W.; Tong, Y.; Chung, S.K.; Liu, K.J.; Shen, J. Caveolin-1 regulates nitric oxide-mediated matrix metalloproteinases activity and blood–brain barrier permeability in focal cerebral ischemia and reperfusion injury. J. Neurochem. 2012, 120, 147–156. [Google Scholar] [CrossRef] [PubMed]
  197. Chandler, S.; Miller, K.M.; Clements, J.M.; Lury, J.; Corkill, D.; Anthony, D.C.C.; Adams, S.E.; Gearing, A.J.H. Matrix metalloproteinases, tumor necrosis factor and multiple sclerosis: An overview. J. Neuroimmunol. 1997, 72, 155–161. [Google Scholar] [CrossRef]
  198. Inzitari, D.; Giusti, B.; Nencini, P.; Gori, A.M.; Nesi, M.; Palumbo, V.; Piccardi, B.; Armillis, A.; Pracucci, G.; Bono, G.; et al. MMP9 Variation After Thrombolysis Is Associated with Hemorrhagic Transformation of Lesion and Death. Stroke 2013, 44, 2901–2903. [Google Scholar] [CrossRef] [PubMed]
  199. Montaner, J.; Molina, C.A.; Monasterio, J.; Abilleira, S.; Arenillas, J.F.; Ribo, M.; Quintana, M.; Alvarez-Sabin, J. Matrix Metalloproteinase-9 Pretreatment Level Predicts Intracranial Hemorrhagic Complications After Thrombolysis in Human Stroke. Circulation 2003, 107, 598–603. [Google Scholar] [CrossRef] [Green Version]
  200. Arba, F.; Piccardi, B.; Palumbo, V.; Giusti, B.; Nencini, P.; Gori, A.M.; Sereni, A.; Nesi, M.; Pracucci, G.; Bono, G.; et al. Small Vessel Disease Is Associated with Tissue Inhibitor of Matrix Metalloproteinase-4 After Ischaemic Stroke. Transl. Stroke Res. 2018, 10, 44–51. [Google Scholar] [CrossRef]
  201. Candelario-Jalil, E.; Thompson, J.; Taheri, S.; Grossetete, M.; Adair, J.C.; Edmonds, E.; Prestopnik, J.; Wills, J.; Rosenberg, G.A. Matrix Metalloproteinases Are Associated with Increased Blood–Brain Barrier Opening in Vascular Cognitive Impairment. Stroke 2011, 42, 1345–1350. [Google Scholar] [CrossRef] [Green Version]
  202. Rosenberg, G.A.; Sullivan, N.; Esiri, M.M. White matter damage is associated with matrix metalloproteinases in vascular dementia. Stroke 2001, 32, 1162–1168. [Google Scholar] [CrossRef] [Green Version]
  203. Rosenberg, G.A. Inflammation and white matter damage in vascular cognitive impairment. Stroke 2009, 40, S20–S23. [Google Scholar] [CrossRef] [Green Version]
  204. Ketsawatsomkron, P.; Keen, H.L.; Davis, D.R.; Lu, K.T.; Stump, M.; De Silva, T.M.; Hilzendeger, A.M.; Grobe, J.L.; Faraci, F.M.; Sigmund, C.D. Protective role for tissue inhibitor of Metalloproteinase-4, a novel peroxisome proliferator-activated receptor-γ target gene, in smooth muscle in Deoxycorticosterone acetate-salt hypertension. Hypertension 2016, 67, 214–222. [Google Scholar] [CrossRef] [Green Version]
  205. Radomski, A.; Jurasz, P.; Sanders, E.J.; Overall, C.M.; Bigg, H.F.; Edwards, D.R.; Radomski, M.W. Identification, regulation and role of tissue inhibitor of metalloproteinases-4 (TIMP-4) in human platelets. Br. J. Pharmacol. 2002, 137, 1330–1338. [Google Scholar] [CrossRef] [PubMed]
  206. Tomimoto, H.; Akiguchi, I.; Wakita, H.; Osaki, A.; Hayashi, M.; Yamamoto, Y. Coagulation activation in patients with Binswanger disease. Arch. Neurol. 1999, 56, 1104–1108. [Google Scholar] [CrossRef]
  207. Iwamoto, T.; Kubo, H.; Takasaki, M. Platelet activation in the cerebral circulation in different subtypes of ischaemic stroke and Binswanger’s disease. Stroke 1995, 26, 52–56. [Google Scholar] [CrossRef] [PubMed]
  208. Bjerke, M.; Zetterberg, H.; Edman, A.; Blennow, K.; Wallin, A.; Andreasson, U. Cerebrospinal fluid matrix metalloproteinases and tissue inhibitor of metalloproteinases in combination with subcortical and cortical biomarkers in vascular dementia and Alzheimer’s disease. J. Alzheimers Dis. 2011, 27, 665–676. [Google Scholar] [CrossRef] [PubMed]
  209. Zhang, J.; Liu, N.; Yang, C. Effects of rosuvastatin in combination with nimodipine in patients with mild cognitive impairment caused by cerebral small vessel disease. Panminerva Med. 2019, 61, 439–443. [Google Scholar] [CrossRef]
  210. Moretti, R.; Torre, P.; Antonello, R.M.; Cazzato, G.; Pizzolato, G. Different responses to rivastigmine in subcortical vascular dementia and multi-infarct dementia. Am. J. Alzheimers Dis. Other Demenentias 2008, 23, 167–176. [Google Scholar] [CrossRef]
  211. Laleu, B.; Gaggini, F.; Orchard, M.; Fioraso-Cartier, L.; Cagnon, L.; Houngninou-Molango, S.; Gradia, A.; Duboux, G.; Merlot, C.; Heitz, F.; et al. First in class, potent, and orally bioavailable NADPH oxidase isoform 4 (Nox4) inhibitors for the treatment of idiopathic pulmonary fibrosis. J. Med. Chem. 2010, 53, 7715–7730. [Google Scholar] [CrossRef]
  212. De Silva, T.M.; Miller, A.A. Cerebral Small Vessel Disease: Targeting Oxidative Stress as a Novel Therapeutic Strategy? Front. Pharmacol. 2016, 7, 61. [Google Scholar] [CrossRef] [Green Version]
  213. Pretnar-Oblak, J.; Sebestjen, M.; Sabovic, M. Statin treatment improves cerebral more than systemic endothelial dysfunction in patients with arterial hypertension. Am. J. Hypertens. 2008, 21, 674–678. [Google Scholar] [CrossRef] [Green Version]
  214. Amarenco, P.; Benavente, O.; Goldstein, L.B.; Callahan, A.; Sillesen, H.; Hennerici, M.G.; Gilbert, S.; Rudolph, A.E.; Simunovic, L.; Zivin, J.A.; et al. Stroke Prevention by Aggressive Reduction in CholesterolLevels Investigators. Results of the stroke prevention by aggressive reduction in cholesterol levels (SPARCL) trial by stroke subtypes. Stroke 2009, 40, 1405–1409. [Google Scholar] [CrossRef] [Green Version]
  215. Caruso, G.; Godos, J.; Privitera, A.; Lanza, G.; Castellano, S.; Chillemi, A.; Bruni, O.; Ferri, R.; Caraci, F.; Grosso, G. Phenolic Acids and Prevention of Cognitive Decline: Polyphenols with a Neuroprotective Role in Cognitive Disorders and Alzheimer’s Disease. Nutrients 2022, 14, 819. [Google Scholar] [CrossRef] [PubMed]
  216. Del Rio, D.; Rodriguez-Mateos, A.; Spencer, J.P.E.; Tognolini, M.; Borges, G.; Crozier, A. Dietary (poly)phenolics in human health: Structures, bioavailability, and evidence of protective effects against chronic diseases. Antioxid. Redox Signal. 2013, 18, 1818–1892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Cullen, A.E.; Centner, A.M.; Deitado, R.; Salazar, J.F.A. The Impact of Dietary Supplementation of Whole Foods and Polyphenols on Atherosclerosis. Nutrients 2020, 12, 2069. [Google Scholar] [CrossRef] [PubMed]
  219. Kesse-Guyot, E.; Fezeu, L.; Andreeva, V.A.; Touvier, M.; Scalbert, A.; Hercberg, S.; Galan, P. Total and specific polyphenol intakes in midlife are associated with cognitive function measured 13 years later. J. Nutr. 2012, 142, 76–83. [Google Scholar] [CrossRef]
  220. Goni, L.; Fernández-Matarrubia, M.; Romanos-Nanclares, A.; Razquin, C.; Ruiz-Canela, M.; Martínez-González, M.Á.; Toledo, E. Polyphenol intake and cognitive decline in the Seguimiento Universidad de Navarra (SUN) Project. Br. J. Nutr. 2021, 126, 43–52. [Google Scholar] [CrossRef]
  221. Shakoor, H.; Feehan, J.; Apostolopoulos, V.; Platat, C.; Al Dhaheri, A.S.; Ali, H.I.; Ismail, L.C.; Bosevski, M.; Stojanovska, L. Immunomodulatory Effects of Dietary Polyphenols. Nutrients 2021, 13, 728. [Google Scholar] [CrossRef]
  222. Godos, J.; Caraci, F.; Micek, A.; Castellano, S.; D’Amico, E.; Paladino, N.; Ferri, R.; Galvano, F.; Grosso, G. Dietary Phenolic Acids and Their Major Food Sources Are Associated with Cognitive Status in Older Italian Adults. Antioxidants 2021, 10, 700. [Google Scholar] [CrossRef]
  223. Ran, L.S.; Liu, W.H.; Fang, Y.Y.; Xu, S.B.; Li, J.; Luo, X.; Pan, D.J.; Wang, M.H.; Wang, W. Alcohol, coffee and tea intake and the risk of cognitive deficits: A dose-response meta-analysis. Epidemiol. Psychiatr. Sci. 2021, 30, e13. [Google Scholar] [CrossRef]
  224. Mallik, S.B.; Mudgal, J.; Nampoothiri, M.; Hall, S.; Dukie, S.A.; Grant, G.; Rao, C.M.; Arora, D. Caffeic acid attenuates lipopolysaccharide-induced sickness behaviour and neuroinflammation in mice. Neurosci. Lett. 2016, 632, 218–223. [Google Scholar] [CrossRef]
  225. Lee, A.Y.; Wu, T.T.; Hwang, B.R.; Lee, J.; Lee, M.-H.; Lee, S.; Cho, E.J. The Neuro-Protective Effect of the Methanolic Extract of Perilla frutescens var. japonicaand Rosmarinic Acid against H2O2-Induced Oxidative Stress in C6 Glial Cells. Biomol. Ther. 2016, 24, 338–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. De Mello Andrade, J.M.; Dos Santos Passos, C.; Kieling Rubio, M.A.; Mendonça, J.N.; Lopes, N.P.; Henriques, A.T. Combining in vitro and in silico approaches to evaluate the multifunctional profile of rosmarinic acid from Blechnum brasiliense on targets related to neurodegeneration. Chem. Biol. Interact. 2016, 254, 135–145. [Google Scholar] [CrossRef] [PubMed]
  227. Rahbardar, M.G.; Amin, B.; Mehri, S.; Mirnajafi-Zadeh, S.J.; Hosseinzadeh, H. Anti-inflammatory effects of ethanolic extract of Rosmarinus officinalis L. and rosmarinic acid in a rat model of neuropathic pain. Biomed. Pharmacother. 2017, 86, 441–449. [Google Scholar] [CrossRef] [PubMed]
  228. Dragomanova, S.; Pavlov, S.; Marinova, D.; Hodzev, Y.; Petralia, M.C.; Fagone, P.; Nicoletti, F.; Lazarova, M.; Tzvetanova, E.; Alexandrova, A.; et al. Neuroprotective Effects of Myrtenal in an Experimental Model of Dementia Induced in Rats. Antioxidants 2022, 11, 374. [Google Scholar] [CrossRef] [PubMed]
  229. Lanza, G.; Bramanti, P.; Cantone, M.; Pennisi, M.; Pennisi, G.; Bella, R. Vascular Cognitive Impairment through the Looking Glass of Transcranial Magnetic Stimulation. Behav. Neurol. 2017, 2017, 1421326. [Google Scholar] [CrossRef]
Figure 1. The vicious circle of SVD pathology.
Figure 1. The vicious circle of SVD pathology.
Ijms 23 03508 g001
Figure 2. A synopsis of the metalloproteinases action inside the brain, redirecting in SVD pathology.
Figure 2. A synopsis of the metalloproteinases action inside the brain, redirecting in SVD pathology.
Ijms 23 03508 g002
Scheme 1. Synopsis of the pathophysiology of SVD.
Scheme 1. Synopsis of the pathophysiology of SVD.
Ijms 23 03508 sch001
Table 1. Possible hematic or CSF markers of SVD.
Table 1. Possible hematic or CSF markers of SVD.
Functional
Domain
MarkersEffectiveness on SVD
BBB LeakageDCE-MRI technique:
Increase in permeability surface area
Increase in white matter alterations
Lower blood plasma volume in white matter altered regions
Demonstrate diagnostic confirmation
Demonstrate the amount and the progression of SVD
Determine the PS increasing together with a lowering of blood vP
Loss of pericytes: upregulation of FGFBP1 and ANGPT2Altered angiogenesis and demonstration of a venous-shifted molecular pattern of BBB, due to the altered arterial regulatory properties
Enlargement of PVSAlteration of the glymphatic system
M1 activation: increase in TNF-alpha, Il-23, IL-1 beta, and IL-12Strong and chronic neuroinflammatory condition, shifted to a M1 vs. M2 activation
General increment in caspase-3 RNA; of MMP-2Promoting and overwhelming the active neuroinflammation condition
Endothelial dysfunctionDecrease in ENOV, prostacyclins, NO, eNOS, and VE-cadherinsAltered production of NO, due to decrease in its production and increment in its consumption, due to increment in ROS
Increase in C-protein, EDHF, VEGF, ICAM-1, sTM, Il-6, PA-1, von Willebrand Factors, HIF-1 alpha; VEGFR, and NeuroglobinExpression of endothelial altered activation, with important flawless permeability and activation of thrombotic pattern
Increase in homocysteineEndothelial toxicity, promotion of oxidative and inflammatory damages
Increase in CSF/plasma albumin ratioProof of endothelial altered permeability
albuminuriaIndirect proof of endothelial altered permeability
Oxidative damageIncrease in SOD, prostacyclin, and Hydrogen peroxideAltered response to oxidative stress, with damages to mitochondria, altered oxygen delivery, and endothelial degeneration promotion
Decrease in NOX2 NADPH oxidaseFurther reduction of proper response to ROS accumulation; their decrease is proportional to endothelial inflammation and alteration
APOE4Promotion of endothelial reduced resistance to ROS
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Moretti, R.; Caruso, P. Small Vessel Disease: Ancient Description, Novel Biomarkers. Int. J. Mol. Sci. 2022, 23, 3508. https://doi.org/10.3390/ijms23073508

AMA Style

Moretti R, Caruso P. Small Vessel Disease: Ancient Description, Novel Biomarkers. International Journal of Molecular Sciences. 2022; 23(7):3508. https://doi.org/10.3390/ijms23073508

Chicago/Turabian Style

Moretti, Rita, and Paola Caruso. 2022. "Small Vessel Disease: Ancient Description, Novel Biomarkers" International Journal of Molecular Sciences 23, no. 7: 3508. https://doi.org/10.3390/ijms23073508

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

Article Metrics

Back to TopTop