9
This article appeared in a journal published by Elsevier. The attached copy is furnished to the author for internal non-commercial research and education use, including for instruction at the authors institution and sharing with colleagues. Other uses, including reproduction and distribution, or selling or licensing copies, or posting to personal, institutional or third party websites are prohibited. In most cases authors are permitted to post their version of the article (e.g. in Word or Tex form) to their personal website or institutional repository. Authors requiring further information regarding Elsevier’s archiving and manuscript policies are encouraged to visit: http://www.elsevier.com/copyright

Blood-brain barrier alterations in ageing and dementia

Embed Size (px)

Citation preview

This article appeared in a journal published by Elsevier. The attachedcopy is furnished to the author for internal non-commercial researchand education use, including for instruction at the authors institution

and sharing with colleagues.

Other uses, including reproduction and distribution, or selling orlicensing copies, or posting to personal, institutional or third party

websites are prohibited.

In most cases authors are permitted to post their version of thearticle (e.g. in Word or Tex form) to their personal website orinstitutional repository. Authors requiring further information

regarding Elsevier’s archiving and manuscript policies areencouraged to visit:

http://www.elsevier.com/copyright

Author's personal copy

Blood-brain barrier alterations in ageing and dementia

Bogdan O. Popescu a,b,⁎, Emil C. Toescu c, Laurenţiu M. Popescu a, Ovidiu Bajenaru b, Dafin F. Muresanu d,Marianne Schultzberg e, Nenad Bogdanovic f

a Laboratory of Molecular Medicine, ‘Victor Babeş’ National Institute of Pathology, Bucharest 050096, Romaniab Department of Neurology, University Hospital Bucharest, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest 050098, Romaniac Department of Physiology, Division of Medical Sciences, The Medical School, University of Birmingham, Birmingham B15 2TT, UKd Department of Neurology, ‘Iuliu Hatieganu’ University of Medicine and Pharmacy, Cluj Napoca 400023, Romaniae Division of Neurodegeneration, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm SE-141 86, Swedenf Alzheimer's Disease Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm SE-141 86, Sweden

a b s t r a c ta r t i c l e i n f o

Available online 4 March 2009

Keywords:AgeingAlzheimer's diseaseBlood-brain barrierBrain capillariesMechanisms of neurodegenerationNeurovascular couplingVascular dementiaParkinson's diseaseTrophic factors

The current pathogenic scenarios of different types of dementia are based on a number of commonmechanisms of neurodegeneration, such as accumulation of abnormal proteins (within or outside cells),mitochondrial dysfunction and oxidative stress, calcium homeostasis dysregulation, early synapticdisconnection and late apoptotic cell death. Ageing itself is associated with mild cognitive deterioration,probably due to subtle multifactorial changes resulting in a global decrease of a functional brain reserve.Increased age is a risk factor for neurodegeneration and key pathological features of dementia can also befound in aged brains. One of the underexplored brain structures in ageing and dementia is the blood-brainbarrier (BBB), a complex cellular gate which regulates tightly the transport of molecules into and from thecentral nervous system. Disruption of this barrier is now increasingly documented not only in brain vasculardisease but also in ageing and neurodegenerative disorders. To date, such evidence points mainly at anassociation between various dementia forms and disruption of the BBB. But, in reviewing such results, andtaking into account the exquisite sensitivity of neuronal function to the composition of the interstitial brainfluid (IBF), which is regulated by the BBB, we would like to propose the existence of a possible causal linkbetween alterations of BBB and conditions associated with cognitive decline.

© 2009 Elsevier B.V. All rights reserved.

1. Introduction

In order to function properly, the central nervous system (CNS),and the neurones in particular, require the maintenance of ionicconcentrations (Na+, K+ and Ca2+) within narrow ranges and ofadequate metabolic substrates. In addition, the neural function is alsovery sensitive to the effects of a wide variety of circulating substances(both proteins and non-proteins) that are not harmful to theperipheral organs. Such strict separation between the neural millieuand the circulatory space is achieved through the properties of aunique biological structure, essentially composed of capillaryendothelial cells, basal lamina and astrocytes, and acting as a two-way diffusion barrier — the blood-brain barrier (BBB). Pericytes, theleast studied BBB constituents, are mesenchymal-like cells surround-ing the endothelial cells and able to contract brain capillaries and tochange the blood flow [1,2]. The peculiarity of BBB (Fig. 1) is given, atleast in part, by a specific phenotype of the endothelial cells, with

adherens junctions as cell–cell interaction stabilizers, and tightjunctions (TJs) that limit the paracellular flow of water, ions andlarger molecules into the brain (gate function) and organize the cellmembrane in apical–basal domains (fence function) [3]. The ‘sealing’feature of the TJs is conferred by the expression in endothelial cells oftransmembrane proteins such as occludin [4], claudins [5] andjunction adhesion molecules [6], anchored via accessory proteins tothe cytoskeleton [7]. This ‘BBB phenotype’ of endothelial cells ischaracterized by a low pinocytic activity and is also much moreeffective in limiting the paracellular flow of ions as compared to that inperipheral capillaries, as reflected by a ~100 fold higher transen-dothelial electrical resistance in the CNS [8]. Therefore, the moleculartraffic at the BBB level takes place mostly by transcellular means.Gases, such as O2 and CO2, are exchanged through the lipidmembranes, as a result of gradient diffusion, while the influx ofamino acids and glucose is regulated through specific transporters.Larger molecules such as peptides (e.g. leptin, insulin, and transferrin)are transported by receptor-mediated transcytosis. With the excep-tion of the receptor-guided molecules, passage of all other hydrophiliccompounds, including drugs, is highly restricted under normalconditions. Size generally represents another passage criterion,substances with a molecular weight higher than 500 Da being unable

Journal of the Neurological Sciences 283 (2009) 99–106

⁎ Corresponding author. Laboratory of Molecular Medicine, ‘Victor Babeş’ NationalInstitute of Pathology, Bucharest 050096, Romania. Tel.: +40 744 35 34 33; fax: +40 21312 81 02.

E-mail address: [email protected] (B.O. Popescu).

0022-510X/$ – see front matter © 2009 Elsevier B.V. All rights reserved.doi:10.1016/j.jns.2009.02.321

Contents lists available at ScienceDirect

Journal of the Neurological Sciences

j ourna l homepage: www.e lsev ie r.com/ locate / jns

Author's personal copy

to cross the BBB [7]. Some of the transporters are specialized forendothelial efflux, for instance P-glycoprotein, which acts as an active,ATP-dependent pump [9]. Water flow into and out of the brain tissueis regulated by aquaporins (AQP), AQP-4 being expressed in astrocyteperivascular endfeet [10] to form water channels and to enable fluidhomeostasis in the brain. In contrast, the brain endothelial cells do notexpress AQP channels [11]. Besides the physical and transport barriersdescribed above, an enzymatic barrier made of nucleotidases andpeptidases is active in the proximity of the capillaries [12].

Another potentially important theoretical issue is the physiologicalregulation of BBB permeability. The current view is that under normalcircumstances the tightness and transport properties of the BBB arerelatively constant, and this is in agreement with the need for aconserved extracellular milieu allowing normal CNS functioning.However, the major features of the endothelial cell phenotype in theBBB are dependent on astrocytes [13], indicated by the fact thatprotein expression and distribution in endothelia are induced byastrocyte-derived signals, and thereby being adaptable. In mixedculture conditions for instance, astrocytes induce the differentiation ofendothelial cells and pericytes partly via secretion of transforminggrowth factor beta 1 (TGFβ1) [14], which also alters the expression ofbasement membrane-related molecules [15]. Moreover, there isevidence showing that other growth factors synthesized and secretedby astrocytes, such as the vascular endothelial growth factor (VEGF)[16,17], the basic fibroblast growth factor (bFGF) [17,18] and the glialcell line-derived neurotrophic factor (GDNF) [19], are involved inmodulation of BBB permeability. Recent reports also suggested thatcalcium signals originating in astrocytes can be transferred to capillary

endothelial cells and mediate BBB transport processes [20]. Mobiliza-tion of cytosolic calcium (Ca2+) in astrocytic endfeet occurs forinstance as a result of purinergic receptors (P2Y) signalling; however,how endothelial cells respond to these Ca2+ waves is not elucidated[21]. Endothelial cells bear also receptors of tumour necrosis factor-α(TNF-α) receptor superfamily, such as TNFR1, Fas and DR5, but theirligands do not trigger cell death in this cell type. Instead, they are ableto upregulate chemokines and adhesion molecules and to activate theextracellular signal regulated kinases [22]. Besides, TNF-α can inducean upregulation of the efflux transporter P-glycoprotein in endothelialcells [23]. However, the regulation of occludin and claudins inendothelial cells and the factors inducing their low pinocyticphenotype, key features of the BBB gate function maintenance, arenot yet clarified.

2. Ageing impact on blood-brain barrier

Dysfunction of the BBB in the ageing brain was documented byvarious studies and is at least in part responsible for pathologicalalterations such as white matter lesions [24], which in turn correlatewith progressive cognitive deterioration [25]. In a recent review,Farrall and Wardlaw extensively document the increase in BBBpermeability with ageing in healthy individuals. The commonly usedmethods to analyze the BBB function in studies in humans are eitherlinked to brain imaging or biochemical. By means of assessment of thecerebrospinal fluid (CSF)/plasma albumin ratio, different studiesdemonstrated significantly higher albumin leakage through the BBB inold healthy as compared to young healthy individuals, and these

Fig. 1. Schematic representation of the blood-brain barrier (BBB) structure, transport and regulation. The capillaries and the brain tissue are separated by a barrier formed byendothelial cells, pericytes and astrocyte end-feet. Endothelial cells are sealed by tight and adherens junctions. The tight junctions are composed of transmembrane proteins such asoccludin, claudins and junction adhesionmolecules (JAM), which grasp actin-anchored cytoplasmic proteins such as ZO1, ZO2 and ZO3. Transport through the BBBmostly takes placeby transcellular means: diffusion of gases (O2, CO2), transporters (e.g. GLUT-1 for glucose), or receptor-mediated transcytosis (e.g. insulin). P-glycoprotein (P-gp) operates theendothelial efflux of different compounds, including drugs, and aquaporin 4 (AQP-4)modulates thewater flow into the brain. Physiological regulation of BBB permeability is probablyregulated by both blood-borne and astrocytic factors, such as growth factors (transforming growth factor beta 1 — TGFβ1, vascular endothelial growth factor — VEGF, the basicfibroblast growth factor— bFGF, glial cell line-derived neurotrophic factor— GDNF), tumour necrosis factor (TNF) family members and calcium signals. Extracellular signal-regulatedkinases (ERK) are activated through TNF receptors and are able to influence transcription via transcription factors. In BBB endothelial cells, localization of many proteins at theluminal and abluminal membranes differs, thus creating a ‘polarization’ (e.g. P-gp is expressed only in luminal capillary membranes; GLUT-1 is present at both cellular versants butasymmetrically). Moreover, there are proteins important for BBB transport expressed only in astrocytes, and not in endothelial cells, and vice versa (e.g. AQP-4 is abundant in glialendfeet but is absent in endothelial cells). Mobilization of cytosolic calcium (Ca2+) in astrocytic endfeet occurs as a result of purinergic receptors (P2Y) signalling; however, howendothelial cells respond to these Ca2+ waves is not elucidated.

100 B.O. Popescu et al. / Journal of the Neurological Sciences 283 (2009) 99–106

Author's personal copy

results were also confirmed by studies using brain imaging (computedtomography — CT, magnetic resonance imaging — MRI or positron-emission tomography — PET) [26].

In a rodent model of senescence, morphological changes of BBB[27] and leakage of endogenous albumin [28] and IgG [29] to the brainparenchymawere selectively demonstrated for brain regions involvedin cognition, such as the hippocampus. The senescence-acceleratedprone mouse strain 8 (SAMP8), which shows age-related learning andmemory deficits [27], exhibits increased oxidative stress at an earlyage [30], before other pathological features are reported. Decreasedlevels of superoxide dismutase [31,32] and glutathione peroxidase[33] seem to be responsible for this modification. An age-relatedincrease in amyloid precursor protein (APP) expression withoutplaque formation was described in the brain of the SAMP8 mice [34].These results suggest that oxidative stress in brain parenchyma cantrigger alterations of the BBB, possibly by cell death, gliosis andsignalling changes. Moreover, in aged Wistar rats, which showimpairment in short term-memory, leakage through the BBB wasfound to be associated with microglial activation [35]. The latter is asource of oxidative damage [36] possibly not only to neurones, butalso to glial and endothelial cells. Leakage of BBB can inducemicroglialactivation by letting abnormal molecules pass into the brainparenchyma and in turn free radicals released from the microgliamay further alter the BBB, in a vicious cycle. In addition, passage ofneuroimmune factors to the brain also changes with senescence.Furthermore, increased transport of TNF-α has been shown in somebrain regions of old SAMP8 mice, suggesting that beyond modulatingBBB properties, TNF-α is able to cross the BBB more efficiently withage [37], while passage of interleukin-1-β (IL-1β) through the BBBdecreases with age [38]. It was also suggested that the aged BBB is notable to transfer glucose properly to the brain, since expression of theglucose transporter GLUT-1 is reduced, both in physiologically agedand in SAMP8 mice [39]. Neurotrophin-like peptides are able toreverse this modification [40], suggesting that protein expression atthe BBB level could be influenced by neurotrophines during ageing.

Another effect of brain ageing is the accumulation of iron inastrocytes [41] and one suggested mechanism for this is the reductionof ceruloplasmin expression in the CNS [42]. Iron in excess cangenerate free radicals and harm cells [43], therefore its increase in theperivascular astrocytes could participate in alterations seen in BBBwith ageing. Transferrin mRNA levels have been shown to increase inthe brainwith age, independent to blood-borne factors [44]. However,the reverse scenario, i.e. an increased passage of iron to the brain dueto improper barrier function, cannot be excluded [45].

A different mechanism for BBB dysfunction in the elderly is thedecreased activity of the efflux transporter P-glycoprotein that mayresult in decreased extrusion of toxins from the brain to capillaries.Toornvliet and coauthorsmeasured P-glycoprotein activity inyoung andelderly healthy volunteers by (R)-[(11)C] verapamil andPETand found asignificantly increased grey substance distribution for the ligand in theaged subjects [46], suggesting a decreased P-glycoprotein activity.

A decline in estrogen levels may also be involved in age-relatedmalfunction of BBB and may result in sensitization to neurodegenera-tion. Thus, Bake and Sohrabji found that BBB leakage is 2 to 4 timesincreased in senescent ovariectomized rats as compared to the youngadult controls. Surprisingly, estrogen replacement resulted in a furtherincrease of leakage at the hippocampal level [47]. Moreover, a recentstudy showed that mid-life adiposity factors correlate with BBBleakage in late life and suggested a sex-hormone mediated mechan-ism for this alteration [48]. In turn, an altered transfer through theaged BBB of leptin [49], a hormone of adipose tissue origin linked toobesity, could induce its deleterious central effects, which may bereversible [50]. Furthermore, ageing and obesity are both associatedwith reduced sensitivity to insulin in the brain [51,52], which candecrease the clearance of β-amyloid (Aβ) peptide by a mechanismlinked to BBB [53].

3. Blood-brain barrier in cerebrovascular disease and vasculardementia

Vascular dementia (VD) is a syndrome with different etiologies anddifferent pathogenic mechanisms, which associates with some commonvascular risk factors [54]. Such a risk factor, perhaps the most important,is arterial hypertension. In epidemiological studies, antihypertensivesshowed a robust effect as protectors against development of dementia[55]. It was hypothesized that changes in brain microvasculature inresponse to chronic hypertension, for instance lipohyalinosis andfibrosis,may induce BBB failure, which further contributes to lacunar strokes,leukoaraiosis and dementia [56]. Indeed, a recent report showed thathypertension can induce BBB alterations by modulation of proteinexpression level. In this study, spontaneously hypertensive rats (SHR)displayed lower levels of the TJ protein zonula occludens-2 (ZO-2) andhigher levels of the ion transporter, Na+/H+exchanger 1, as compared tocontrol rats whichmaintained normal blood pressure values. In contrast,the expression of ZO-1, occludin, actin, claudin-5 in endothelial cells atthe BBB level remained unchanged [57]. Themost acute and threateningeffects of hypertension on BBB may occur in hypertension encephalo-pathy, a condition inwhich the cerebral perfusion is elevated beyond thelimits of autoregulation. In Dahl salt-sensitive rats, the most commonlyusedmodel forhypertensionencephalopathy, BBB leakage ismassive andcan be reversed by treatment with δ V1-1, a selective peptide inhibitor ofδ protein kinase C (δPKC) [58,59]. Therefore, δPKC could be a promisingtherapeutic target for theprotection of BBBunder vascular stress [60] andpossibly for the prevention of cognitive impairment in cardiovascularpatients.OtherPKC inhibitorswere alsopreviously reported to reduce theendothelial permeability induced by deleterious factors such as oxidativestress [61,62] and therefore it can be claimed that basic cell signallingmechanisms regulate endothelial transfer properties at least in patho-logical conditions. Besides kinase inhibitors, magnesium sulphate hasbeen shown to reduce BBB permeability and brain oedema in an animalmodel for eclampsia by an AQP-4-independent mechanism [63]. More-over, hypertension is not only able to alter BBB properties per se, but canalso aggravate the BBB leakage and cognitive decline triggered byenvironmental factors, such as heat stress [64].

Type 2 diabetes mellitus appears to be a risk factor for developingVD, particularly if treated with insulin [65,66]. Despite being aperipheral disease, diabetes is able to induce alterations in BBBpermeability [67], and this may be a mechanism for its role in VDcaused by small vessels disease. In a rat model of diabetes it has beenshown that the expression of GLUT-1 and GLUT-3 is downregulated,probably by an adaptive mechanism of protection against sustainedhigh blood glucose levels [68]. Furthermore, in an in vitro BBB modeland in streptozotocin-induced diabetic rats, the efflux transporter P-glycoprotein was found to be upregulated by high glucose through aNF-κB-dependent mechanism [69]. Hawkins and colleagues showedthat in experimental rat diabetes BBB permeability to sucroseincreased in parallel to a decrease in the level of the TJs proteinsoccludin and ZO-1 [70]. Interestingly, occludin expression in neuronesis elevated in VD brains, suggesting an additional biological role forthis protein [71]. Moreover, a recent study showed that in diabetic ratsthe BBB permeability is progressively increased despite normalizationof blood glucose levels by insulin treatment [72]. Based on themolecular weight passage pattern, the authors suggested that the BBBfailure in diabetes mellitus is mainly due to a loosening of theparacellular path. Besides affecting the properties of endothelial cells,diabetes may act on astrocyte phenotype, which is the otherimportant regulator of molecular transport of the BBB [73].

High cholesterol and dyslipidemia are well-known vascular riskfactors that associate with cognitive decline, stroke, VD and Alzheimer'sdisease (AD), even though the molecular basis needs clarifying [74].Brain cholesterol represents a quarter of the total cholesterol in thehuman body. It is synthesized locally and exchange with circulatingcholesterol is normally prevented by the BBB [75]. The main carrier of

101B.O. Popescu et al. / Journal of the Neurological Sciences 283 (2009) 99–106

Author's personal copy

brain cholesterol is apolipoprotein E (apoE),whichensures a continuousmolecular exchange between astrocytes and neurones [76]. However, aconversion of brain cholesterol to 24 S-hydroxycholesterol occurs bymediation of the neuronal cytochrome P-450 species CYP46 and thisform can pass the BBB and be extruded from the brain [77]. In animalmodels, high blood cholesterol levels have been shown to affect thebrain and the BBB. Thus, a long-term cholesterol-enriched diet triggeredBBB disruption and increased the iron deposition and Aβ accumulationin rabbit brains [78]. Furthermore, long-term cholesterol-enriched dietin mice resulted in a marked activation of astrocytes and microgliaincluding an increased expression of inflammatory proteins such ascaspase-1 and the cytokine interleukin-6 (IL-6) [79,80]. In addition, thecholesterol-enriched diet resulted in increased expression of the anti-oxidant protein NAD(P)H:quinone oxidoreductase (NQO1), which waspotentiated in apoE knockout (KO) mice [79], as well as in neuronalaccumulation of hyperphosphorylated tau protein in apoE KOmice [81].Statins, inhibitors of cholesterol synthesis, have been showed to providea protective effect on endothelial permeability in diabetic rats, however,apparently not via inhibition of the cholesterol synthesizing enzyme 3-hydroxy-3-methylglutarylcoenzyme A (HMGCoA), but through a directaction on the endothelium [82]. The protective effect of statins on BBBmay be explained by a decreased interaction of monocytes with theendothelial cells through inhibition of the phosphoinositide 3 kinase/Akt pathway [83].

Stroke is one of the events potentially leading to VD. Cerebralischemia is known to cause increased intracellular calcium levels andexcitotoxicity for neurones, as well as for endothelial cells [84]. Studieson in vitro experimental models, such as hypoxia/aglycemia ofendothelial cells, showed profoundly altered transendothelial elec-trical resistance, an effect that was reversible upon blocking thecalcium channels [85]. Furthermore, it was suggested that themechanism of BBB disruption in stroke is due to the alteration inadherens and tight junction function through modulation of proteinexpression, induced by calcium dyshomeostasis [86]. Moreover,factors released by endothelial cells, such as endothelin-1 and nitricoxide (NO), contribute to changes in the brain microvasculature inboth hypertension and stroke [87] and these changes occur bothbefore and after the ischemic stroke [88]. In this line of evidence,postischemic reperfusion was shown to alter the BBB structure andpermeability [89]. Not only brain ischemia, but also hemorrhagic

stroke seems to be preceded by alterations in BBB properties. In arecent report, Lee and colleagues brought evidence that focallyincreased brain permeability in the SHR model, as demonstrated byMRI, occurred 1 to 2 weeks before the intracerebral hemorrhage [90].

4. Blood-brain barrier in Alzheimer's disease

Even though there is no unifying theory that is able to fully explainthe neuropathological lesions and the progression of the neurodegen-erative process in AD, this disease is still best defined by dementiawitha peculiar accumulation of Aβ peptide and hyperphosphorylated tau inthe brain [91]. The ApoE genotype is a known factor for AD [92], andinflammation [93], vascular factors [94], oxidative stress and synapticdysfunction [95], alterations in cell signalling [96] and sensitization tocell death [97] all appear to be important pathogenic mechanisms.Many of these factors are encountered in other dementia types,mainlyin VD, therefore giving rationale for the concept that most of thedementia cases in fact have amixed origin [54]. The importance of BBBalterations in AD is well established [98] and it is worthy to notice thatmany of the above mentioned disease mechanisms also affect the BBB(e.g. oxidative stress, vascular factors, inflammation). Furthermore,one crucial link between ageing and AD can be an alteredmanagementof Aβ at the BBB level [99]. First, since Aβ is produced not only in thebrain but also in the periphery, leakage of Aβ from capillaries to thebrain may occur both in aged and in pathologically altered BBB.Moreover, it was suggested that the receptor for advanced glycationend products (RAGE), which mediates transfer of Aβ to the brainthrough the endothelial cells [100], can be upregulated during ageing[101]. Second, a diminished extrusion of Aβ from the brain occursduring ageing, by a decreased low-density lipoprotein (LDL) receptor-related protein (LRP)-mediated endothelial transcytosis of Aβ [102].Moreover, in P-glycoprotein null mice, clearance of Aβ from the brainfalls by 50%, suggesting that not only LRP, but also P-glycoprotein isimportant in Aβ clearance [103]. As already mentioned, the activity ofP-glycoprotein decreases with age [46] and therefore excessiveaccumulation of Aβ due to BBB impairment could be one of the linksbetween ageing and AD. Indeed, a proof that BBB alteration precedesaccumulation of senile plaques came from work performed in ADtransgenic mice, in which a breakdown of BBB was demonstrablebefore Aβ deposits were seen in the brain [104]. In current therapeutic

Table 1Molecular mechanisms associated with BBB disruption in ageing and different dementia types and proposed protective treatments.

Mechanism Possible intervention Ageing or disease Model/human References

Increased oxidative stress Antioxidants, gene therapy to target SOD Ageing SAMP 8 mice [30–33]Microglia activation Anti-inflammatory drugs Ageing Physiologically aged rats [35]Increased transport of TNF throughBBB

Anti-TNF antibodies Ageing SAMP 8 mice [37]

Decreased expression of GLUT-1 Neurotrophines, neurotrophic-likepeptides

Ageing, diabetes (risk factor for VD) Physiologically aged and SAMP8 mice, SDR

[39,40,68]

Iron accumulation Iron chelators Ageing, high cholesterol (risk factor for VD), PD Human, physiologically aged mice,cholesterol-enriched rabbits

[41,42,78,119]

Decreased activity of P-gp Gene therapy targeting P-gp Ageing, AD, PD Human, P-gp null transgenic mice [46,103,118]Decreased estrogen level New methods of estrogen replacement Ageing Senescent ovariectomized rats [47]Reduced sensitivity to insulin Insulin, IGF-1 Ageing Human, physiologically aged mice [51,52]Decreased occludin expression Insulin, gene therapy, statins Diabetes (risk factor for VD) SDR [70]Decreased ZO-1 expression Insulin, gene therapy, statins Diabetes (risk factor for VD) SDR [70]Decreased ZO-2 expression Antihypertensives, gene therapy Hypertension (risk factor for both AD and VD) SHR [57]Increased endothelia–monocytesinteraction

Inhibition of PI3K/Akt Diabetes (risk factor for VD) SDR [82,83]

Activation of δPKC δPKC specific inhibitors Hypertension (risk factor for both AD and VD) DSSR [58,59,60]Alteration of adherens and TJsfunction

Calcium blockers Stroke (risk factor for both AD and VD) In vitro BBB model, rats [85,86]

RAGE upregulation RAGE blockers Ageing, AD AD transgenic mice [101]LRP downregulation Gene therapy, other Aβ clearance strategies Ageing, AD AD transgenic mice [102,109]

SOD= superoxide dismutase, SAMP= senescence-accelerated pronemice strain 8, TNF= tumour necrosis factor, BBB= blood-brain barrier, GLUT-1= glucose transporter 1, VD=vascular dementia, SDR= streptozotocin-induced diabetic rats, PD=Parkinson's disease, P-gp= P-glycoprotein, IGF-1= insulin-like growth factor 1, ZO=zonula occludens, SHR=spontaneous hypertensive rats, PI3K = phosphoinositide 3 kinase, PKC = protein kinase C, DSSR = Dahl salt-sensitive rats, RAGE = receptor for advanced glycation end products,LRP = low-density lipoprotein receptor-related protein, Aβ = β-amyloid peptide.

102 B.O. Popescu et al. / Journal of the Neurological Sciences 283 (2009) 99–106

Author's personal copy

approaches it has been shown that antibodies to Aβ can induceclearance of Aβ from the brain, through the BBB [105].

ApoE4 is the main genetic risk factor for sporadic AD [92]. Micedeficient in apoE show a compromised BBB function, demonstrated bya spontaneous leakage of dyes into the brain [106], which progresseswith age [107]. Mulder and colleagues showed that a high-fat diet isan important inducer of both atherosclerosis and BBB disruption inapoE KOmice suggesting that protective isoforms of apoE may protectthe brain from pathological lesions induced by high-fat diet and BBBleakage [108]. Interestingly, apoE KO mice also show a decreasedexpression of LRP receptor [109].

A recent systematic review concluded on the existence of BBBdisruption in patients with AD or VD. The authors reported that BBBpermeability, as assessed by biochemical and imaging methods,significantly increases in both AD and VD as compared to ageingcontrols. Furthermore, the BBB leakage was more pronounced in VDpatients as compared to AD patients [26], and this is in agreementwiththe experimental evidence that vascular factors are able to modify theBBB phenotype and properties (see the previous chapter). Bringing anargument that not only vascular factors could affect the BBB, Algotssonand Winblad recently reported on an altered BBB integrity in asubgroup of AD patients without noticeable vascular lesions [110].Interestingly, menwere primarily affected. However, in a recent reportin Neurology, Bowman and colleagues followed a population of ADpatients for one year and found that BBB impairment is present andconstant in a subgroup of AD individuals, but with no correlationwithage, gender, ApoE genotype or Mini-Mental State examination [111].

It is our view that BBB changes found in AD are not explained only onthebasis of vascular lesions. Thedegeneration of neurones andactivationof glial cells may also affect the endothelial cell phenotype and transportproperties. For instance, BBB disruption has been shown to develop in atransgenic mouse model for taupathies [112], supporting the conceptthat the BBB is affected not only by vascular factors, but also bydegeneration of brain-residing-cells. Dickstein and coworkers have alsoshown that expression of an AD-linked mutation in APP in transgenic

mice is sufficient to compromise the BBB integrity [113]. Moreover,astrocytes and neurones secrete trophic factors such as FGF2 and VEGF[114] that regulate BBB function and can be depleted in dementia [115].Thus, it is conceivable that theBBBalteration inAD isdueboth tovascularfactors and to cell degeneration and abnormal signalling. Moreover,disruption of BBB may accelerate the pathology of the disease, resultingin a vicious cycle. Some of the molecular mechanisms associated withBBB disruption in ageing and different dementia types and proposedprotective treatments are presented in Table 1.

5. Blood-brain barrier in Parkinson's disease

Except for AD and VD, there is little known regarding BBB alter-ations in other neurodegenerative diseases associated with cognitivedecline and dementia. However, a recent review analyses malfunctionof BBB in Parkinson's disease (PD) [98]. Recent data from animalexperiments support a pathogenic link between BBB disruption anddegeneration of dopaminergic neurones. Rite and colleagues foundthat increased permeability by injection of VEGF into the substantianigra resulted in a subsequent loss of dopaminergic neurones [116].Moreover, there is increasing interest in inflammation as a pathogenicmechanism in PD and possibly through alterations in BBB properties,as shown in many different experimental paradigms [117]. The firstevidence of increased permeability of the BBB in PD came from a PETstudy, showing a significantly increased uptake of 11C-verapamil in themidbrain of PD patients relative to controls, suggested to be due to adecreased function of P-glycoprotein [118]. It was also speculated thatan increased BBB passage of metals, such as iron or manganese, maybe involved in PD pathogenesis [119].

6. Conclusions and perspectives

In this review we presented some of the evidence that is rapidlyaccumulating and which implicates dysfunctions of the blood-brainbarrier in a variety of instances connected to cognitive decline, from

Fig. 2. Factors that alter the blood-brain barrier (BBB) and affect cognition. BBB can be disrupted by ageing, vascular factors (hypertension, diabetes, high cholesterol) andinflammation. One important question iswhether BBB lesions could trigger, or at least accelerate, brainpathology, particularlywhen superimposed on a favorable genetic background.Nevertheless, there is data showing that neurodegeneration per se (which associates with e.g. lack of growth factors, Aβ, tangles, inflammatorymediators) can induce BBB disruption.Our hypothesis is that an aggression to any component of the neurovascular unit can ultimately result in brain degeneration, pathological lesions and cognitive decline.

103B.O. Popescu et al. / Journal of the Neurological Sciences 283 (2009) 99–106

Author's personal copy

the normal ageing phenotype to various forms of dementia. Althoughthese observations are currently mainly associational, the fact thatboth in ageing and during early neurodegenerative states theneurones are working in regimens of reduced homeostatic reserve[120] and thus become extremely sensitive and vulnerable to analtered composition of the extracellular milieu, raise the potentiallyvery important question of whether BBB lesions could trigger, or atleast accelerate, brain pathology, particularly when superimposed ona favorable genetic background (Fig. 2). This view was until nowmostly applicable to understanding the etiopathology of multiplesclerosis or neuroinfections, rather than dementia. Nevertheless, thereis increasing data showing that neurodegeneration per se can induceBBB disruption. Therefore, we conclude that an aggression to anycomponent of the neurovascular unit can ultimately result in braindegeneration, pathological lesions and cognitive decline. Moreover, inthe future it may be of interest to target the BBB with specifictherapeutic approaches that could be complementary to strategiesdesigned for neuronal protection or glia.

Acknowledgements

This study has been supported by the Romanian Ministry ofEducation and Research, the National Innovation, Research andDevelopment Plan — grants no 41-013/2007, 61-019-P2/2007, PN06.26-01.01/2007, CEEX 31/2005 and VIASAN 456/2004, by grantsfrom Karolinska Institutet research funds, The Swedish ResearchCouncil and Swedish Brain Power. Our gratefulness is towards thefamilies that donated the brain material for scientific research.

References

[1] Bandopadhyay R, Orte C, Lawrenson JG, Reid AR, De Silva S, Allt G. Contractileproteins in pericytes at the blood-brain and blood-retinal barriers. J Neurocytol2001;30:35–44.

[2] Abbott NJ, Ronnback L, Hannson E. Astrocyte–endothelial interactions at theblood-brain barrier. Nat Rev Neurosci 2006;7:41–53.

[3] Kniesel U, Wolburg H. Tight junctions of the blood-brain barrier. Cell MolNeurobiol 2000;20:57–76.

[4] Furuse M, Hirase T, Itoh M, Nagafuchi A, Yonemura S, Tsukita S. Occludin: a novelintegralmembraneprotein localizing at tight junctions. J Cell Biol 1993;123:1777–88.

[5] Furuse M, Fujita K, Hiiragi T, Fujimoto K, Tsukita S. Claudin-1 and -2: novelintegral membrane proteins localizing at tight junctions with no sequencesimilarity to occludin. J Cell Biol 1998;141:1539–50.

[6] Martìn-Padura I, Lostaglio S, Schneemann M, Williams L, Romano M, Fruscella P,et al. Junctional adhesion molecule, a novel member of the immunoglobulinsuperfamily that distributes at intercellular junctions and modulates monocytetransmigration. J Cell Biol 1998;142:117–27.

[7] Persidsky Y, Ramirez SH, Haorah J, Kanmogne GD. Blood-brain barrier: structuralcomponents and function under physiologic and pathologic conditions. JNeuroimmunol Pharmacol 2006;1:223–36.

[8] Butt AM, Jones HC, Abbott NJ. Electrical resistance across the blood-brain barrierin anaesthetized rats: a developmental study. J Physiol 1990;429:47–62.

[9] Thuerauf N, FrommMF. The role of the transporter P-glycoprotein for dispositionand effects of centrally acting drugs and for the pathogenesis of CNS diseases. EurArch Psychiatry Clin Neurosci 2006;256:281–6.

[10] Nielsen S, Nagelhus EA, Amiry-Moghaddam M, Bourque C, Agre P, Ottersen OP.Specialized membrane domains for water transport in glial cells: high-resolutionimmunogold cytochemistry of aquaporin-4 in rat brain. J Neurosci 1997;17:171–80.

[11] Yool AJ. Aquaporins: multiple roles in the central nervous system. Neuroscientist2007;13:470–85.

[12] Minn A, Ghersi-Egea JF, Perrin R, Leininger B, Siest G. Drug metabolizing enzymesin the brain and cerebral microvessels. Brain Res Brain Res Rev 1991;16:65–82.

[13] Rubin LL, Barbu K, Bard F, Cannon C, Hall DE, Horner H, et al. Differentiation ofbrain endothelial cells in cell culture. Ann N Y Acad Sci 1991;633:420–5.

[14] Ramsauer M, Krause D, Dermietzel R. Angiogenesis of the blood-brain barrier invitro and the function of cerebral pericytes. FASEB J 2002;16:1274–6.

[15] Kose N, Asashima T, Muta M, Iizasa H, Sai Y, Terasaki T, et al. Altered expression ofbasement membrane-related molecules in rat brain pericyte, endothelial, andastrocyte cell lines after transforming growth factor-beta1 treatment. DrugMetabPharmacokinet 2007;22:255–66.

[16] Góra-Kupilas K, Jośko J. The neuroprotective function of vascular endothelialgrowth factor (VEGF). Folia Neuropathol 2005;43:31–9.

[17] Su JJ, Osoegawa M, Matsuoka T, Minohara M, Tanaka M, Ishizu T, et al.Upregulation of vascular growth factors in multiple sclerosis: correlation withMRI findings. J Neurol Sci 2006;243:21–30.

[18] Sobue K, Yamamoto N, Yoneda K, Hodgson ME, Yamashiro K, Tsuruoka N, et al.Induction of blood-brain barrier properties in immortalized bovine brainendothelial cells by astrocytic factors. Neurosci Res 1999;35(2):155–64.

[19] Utsumi H, Chiba H, Kamimura Y, Osanai M, Igarashi Y, Tobioka H, et al. Expressionof GFRalpha-1, receptor for GDNF, in rat brain capillary during postnataldevelopment of the BBB. Am J Physiol Cell Physiol 2000;279:C361–368.

[20] Braet K, Cabooter L, Paemeleire K, Leybaert L. Calcium signal communication inthe central nervous system. Biol Cell 2004;96:79–91.

[21] Simard M, Arcuino G, Takano T, Liu QS, Nedergaard M. Signaling at thegliovascular interface. J Neurosci 2003;23:9254–62.

[22] Wosik K, Biernacki K, Khouzam MP, Prat A. Death receptor expression andfunction at the human blood brain barrier. J Neurol Sci 2007;259:53–60.

[23] Yu C, Kastin AJ, Tu H, Waters S, Pan W. TNF activates P-glycoprotein in cerebralmicrovascular endothelial cells. Cell Physiol Biochem 2007;20:853–8.

[24] Simpson JE, Fernando MS, Clark L, Ince PG, Matthews F, Forster G, et al. Whitematter lesions in an unselected cohort of the elderly: astrocytic, microglial andoligodendrocyte precursor cell responses. Neuropathol Appl Neurobiol2007;33:410–9.

[25] Frisoni GB, Galluzzi S, Pantoni L, Filippi M. The effect of white matter lesions oncognition in the elderly—small but detectable. Nat Clin Pract Neurol 2007;3:620–7.

[26] Farrall AJ, Wardlaw JM. Blood-brain barrier: ageing and microvascular disease —systematic review and meta-analysis. Neurobiol Aging Sep 13 2007 [Electronicpublication ahead of print] PMID: 17869382.

[27] Hosokawa M, Ueno M. Aging of blood-brain barrier and neuronal cells of eye andear in SAM mice. Neurobiol Aging 1999;20:117–23.

[28] Vorbrodt AW, Dobrogowska DH, Ueno M, Tarnawski M. A quantitativeimmunocytochemical study of blood-brain barrier to endogenous albumin incerebral cortex and hippocampus of senescence-accelerated mice (SAM). FoliaHistochem Cytobiol 1995;33:229–37.

[29] Pelegrí C, Canudas AM, del Valle J, Casadesus G, Smith MA, Camins A, et al.Increased permeability of blood-brain barrier on the hippocampus of a murinemodel of senescence. Mech Ageing Dev 2007;128:522–8.

[30] Yasui F, Ishibashi M, Matsugo S, Kojo S, Oomura Y, Sasaki K. Brain lipidhydroperoxide level increases in senescence-accelerated mice at an early age.Neurosci Lett 2003;350:66–8.

[31] Alvarez-García O, Vega-Naredo I, Sierra V, Caballero B, Tomás-Zapico C, Camins A,et al. Elevated oxidative stress in the brain of senescence-accelerated mice at5 months of age. Biogerontology 2006;7:43–52.

[32] Kurokawa T, Asada S, Nishitani S, Hazeki O. Age-related changes in manganesesuperoxide dismutase activity in the cerebral cortex of senescence-acceleratedprone and resistant mouse. Neurosci Lett 2001;298:135–8.

[33] Okatani Y, Wakatsuki A, Reiter RJ, Miyahara Y. Melatonin reduces oxidativedamage of neural lipids and proteins in senescence-acceleratedmouse. NeurobiolAging 2002;23:639–44.

[34] Morley JE, Kumar VB, Bernardo AE, Farr SA, Uezu K, Tumosa N, et al. Beta-amyloidprecursor polypeptide in SAMP8 mice affects learning and memory. Peptides2000;21:1761–7.

[35] Chan-Ling T, Hughes S, Baxter L, Rosinova E, McGregor I, Morcos Y, et al.Inflammation and breakdown of the blood-retinal barrier during “physiologicalaging” in the rat retina: a model for CNS aging. Microcirculation 2007;14:63–76.

[36] Dringen R. Oxidative and antioxidative potential of brain microglial cells.Antioxid Redox Signal 2005;7:1223–33.

[37] Banks WA, Moinuddin A, Morley JE. Regional transport of TNF-alpha across theblood-brain barrier in young ICR and young and aged SAMP8 mice. NeurobiolAging 2001;22:671–6.

[38] McLay RN, Kastin AJ, Zadina JE. Passage of interleukin-1-beta across the blood-brain barrier is reduced in aged mice: a possible mechanism for diminished feverin aging. Neuroimmunomodulation 2000;8:148–53.

[39] Vorbrodt AW, Dobrogowska DH, Meeker HC, Carp RI. Immunogold study ofregional differences in the distribution of glucose transporter (GLUT-1) in mousebrain associated with physiological and accelerated aging and scrapie infection. JNeurocytol Sep 1999;28(9):711–9.

[40] Gschanes A, Boado R, SametzW,WindischM. The drug cerebrolysin and its peptidefraction E021 increase the abundance of the blood-brain barrier GLUT1 glucosetransporter in brains of young and old rats. Histochem J Feb 2000;32(2):71–7.

[41] Connor JR, Menzies SL, St Martin SM, Mufson EJ. Cellular distribution oftransferrin, ferritin, and iron in normal and aged human brains. J Neurosci Res1990;27:595–611.

[42] Jeong SY, David S. Age-related changes in iron homeostasis and cell death in thecerebellum of ceruloplasmin-deficient mice. J Neurosci 2006;26:9810–9.

[43] Aisen P, Enns C, Wessling-Resnick M. Chemistry and biology of eukaryotic ironmetabolism. Int J Biochem Cell Biol 2001;33:940–59.

[44] Moos T, Oates PS, Morgan EH. Expression of transferrin mRNA in rat oligoden-drocytes is iron-independent and changes with increasing age. Nutr Neurosci2001;4:15–23.

[45] Dringen R, Bishop GM, Koeppe M, Dang TN, Robinson SR. The pivotal role ofastrocytes in themetabolism of iron in the brain. NeurochemRes 2007;32:1884–90.

[46] Toornvliet R, van Berckel BN, Luurtsema G, LubberinkM, Geldof AA, Bosch TM, et al.Effect of age on functional P-glycoprotein in theblood-brain barriermeasuredbyuseof (R)-[(11)C]verapamil and positron emission tomography. Clin Pharmacol Ther2006;79:540–8.

[47] Bake S, Sohrabji F. 17beta-estradiol differentially regulates blood-brain barrierpermeability in young and aging female rats. Endocrinology 2004;145:5471–5.

[48] Gustafson DR, Karlsson C, Skoog I, Rosengren L, Lissner L, Blennow K. Mid-lifeadiposity factors relate to blood-brain barrier integrity in late life. J Intern Med2007;262:643–50.

104 B.O. Popescu et al. / Journal of the Neurological Sciences 283 (2009) 99–106

Author's personal copy

[49] Dietrich MO, Spuch C, Antequera D, Rodal I, de Yébenes JG, Molina JA, et al.Megalin mediates the transport of leptin across the blood-CSF barrier. NeurobiolAging Feb 26 2007 [Electronic publication ahead of print] PMID: 17324488.

[50] Banks WA, Farrell CL. Impaired transport of leptin across the blood-brain barrier inobesity is acquired and reversible. Am J Physiol EndocrinolMetab2003;285:E10–15.

[51] Muzumdar RH, Ma X, Yang X, Atzmon G, Barzilai N. Central resistance to theinhibitory effects of leptin on stimulated insulin secretion with aging. NeurobiolAging Sep 2006;27(9):1308–14.

[52] Messier C, Teutenberg K. The role of insulin, insulin growth factor, and insulin-degrading enzyme in brain aging and Alzheimer's disease. Neural Plast 2005;12:311–28.

[53] Shiiki T, Ohtsuki S, Kurihara A, Naganuma H, Nishimura K, Tachikawa M, et al.Brain insulin impairs amyloid-beta(1–40) clearance from the brain. J Neurosci2004;24:9632–7.

[54] Korczyn AD, Vakhapova V. The prevention of the dementia epidemic. J Neurol Sci2007;257:2–4.

[55] Hajjar I, Keown M, Frost B. Antihypertensive agents for aging patients who are atrisk for cognitive dysfunction. Curr Hypertens Rep Dec 2005;7(6):466–73.

[56] Wardlaw JM, Sandercock PA, Dennis MS, Starr J. Is breakdown of the blood-brainbarrier responsible for lacunar stroke, leukoaraiosis, and dementia? Stroke2003;34:806–12.

[57] Hom S, Fleegal MA, Egleton RD, Campos CR, Hawkins BT, Davis TP. Comparativechanges in the blood-brain barrier and cerebral infarction of SHR and WKY rats.Am J Physiol Regul Integr Comp Physiol 2007;292:R1881–1892.

[58] Qi X, Inagaki K, Sobel RA, Mochly-Rosen D. Sustained pharmacological inhibitionof deltaPKC protects against hypertensive encephalopathy through prevention ofblood-brain barrier breakdown in rats. J Clin Invest 2008;118:173–82.

[59] Chou WH, Messing RO. Hypertensive encephalopathy and the blood-brainbarrier: is deltaPKC a gatekeeper? J Clin Invest 2008;118:17–20.

[60] Bright R, Steinberg GK, Mochly-Rosen D. DeltaPKC mediates microcerebrovas-cular dysfunction in acute ischemia and in chronic hypertensive stress in vivo.Brain Res 2007;1144:146–55.

[61] Siflinger-Birnboim A, Johnson A. Protein kinase C modulates pulmonaryendothelial permeability: a paradigm for acute lung injury. Am J Physiol LungCell Mol Physiol 2003;284:L435–451.

[62] Ramírez MM, Kim DD, Durán WN. Protein kinase C modulates microvascularpermeability through nitric oxide synthase. Am J Physiol 1996;271:H1702–1705.

[63] Euser AG, Bullinger L, Cipolla MJ. Magnesium sulphate treatment decreasesblood-brain barrier permeability during acute hypertension in pregnant rats. ExpPhysiol Feb 2008;93(2):254–61.

[64] Muresanu DF, Sharma HS. Chronic hypertension aggravates heat stress inducedcognitive dysfunction and brain pathology: an experimental study in the rat,using growth hormone therapy for possible neuroprotection. Ann N Y Acad Sci2007;1122:1–22.

[65] Ott A, Stolk RP, Hofman A, van Harskamp F, Grobbee DE, Breteler MM. Associationof diabetes mellitus and dementia: the Rotterdam Study. Diabetologia1996;39:1392–7.

[66] Yoshitake T, Kiyohara Y, Kato I, Ohmura T, IwamotoH, Nakayama K, et al. Incidenceand risk factors of vascular dementia and Alzheimer's disease in a defined elderlyJapanese population: the Hisayama Study. Neurology 1995;45:1161–8.

[67] Hawkins BT, Egleton RD. Pathophysiology of the blood-brain barrier: animalmodels and methods. Curr Top Dev Biol 2008;80:277–309.

[68] HouWK, Xian YX, Zhang L, Lai H, Hou XG, Xu YX, et al. Influence of blood glucoseon the expression of glucose trans-porter proteins 1 and 3 in the brain of diabeticrats. Chin Med J (Engl) 2007;120:1704–9.

[69] MaengHJ, KimMH, JinHE, ShinSM, TsuruoT,KimSG, et al. Functional inductionofP-glycoprotein in the blood-brain barrier of streptozotocin-induced diabetic rats:evidence for the involvement of nuclear factor-kappaB, a nitrosative stress-sensitivetranscription factor, in the regulation. Drug Metab Dispos 2007;35:1996–2005.

[70] Hawkins BT, Lundeen TF, Norwood KM, Brooks HL, Egleton RD. Increased blood-brain barrier permeability and altered tight junctions in experimental diabetes inthe rat: contribution of hyperglycaemia and matrix metalloproteinases. Diabe-tologia 2007;50:202–11.

[71] Romanitan MO, Popescu BO, Winblad B, Bajenaru OA, Bogdanovic N. Occludin isoverexpressed in Alzheimer's disease and vascular dementia. J Cell Mol Med2007;11:569–79.

[72] Huber JD, VanGilder RL, Houser KA. Streptozotocin-induced diabetes progres-sively increases blood-brain barrier permeability in specific brain regions in rats.Am J Physiol Heart Circ Physiol 2006;291:H2660–2668.

[73] Savidge TC, Sofroniew MV, Neunlist M. Starring roles for astroglia in barrierpathologies of gut and brain. Lab Invest 2007;87:731–6.

[74] Stella F, Banzato CE, Gasparetto Sé EV, Scudeler JL, Pacheco JL, Kajita RT. Riskfactors for vascular dementia in elderly psychiatric outpatients with preservedcognitive functions. J Neurol Sci 2007;257:247–9.

[75] Björkhem I, Meaney S. Brain cholesterol: long secret life behind a barrier.Arterioscler Thromb Vasc Biol 2004;24:806–15.

[76] Pfrieger FW. Cholesterol homeostasis and function in neurons of the centralnervous system. Cell Mol Life Sci 2003;60:1158–71.

[77] Björkhem I. Crossing the barrier: oxysterols as cholesterol transporters andmetabolic modulators in the brain. J Intern Med 2006;260:493–508.

[78] Ghribi O, Golovko MY, Larsen B, Schrag M, Murphy EJ. Deposition of iron andbeta-amyloid plaques is associated with cortical cellular damage in rabbits fedwith long-term cholesterol-enriched diets. J Neurochem 2006;99:438–49.

[79] Crisby M, Rahman A, Sylvén C, Winblad B, Schultzberg M. Effects of highcholesterol diet on gliosis in apolipoprotein E knockout mice. Implications forAlzheimer's disease and stroke. Neurosci Lett 2004;369:87–92.

[80] Rahman SMA, Van Dam AM, Schultzberg M, Crisby M. High cholesterol dietresults in increased expression of interleukin-6 and caspase-1 in the brain ofapolipoprotein E knockout andwild typemice. J Neuroimmunol 2005;169:59–67.

[81] RahmanA, Akterin S, Flores-Morales A, CrisbyM, KivipeltoM, SchultzbergM, et al.High cholesterol diet induces tau hyperphosphorylation in apolipoprotein Edeficient mice. FEBS Lett 2005;579:6411–6.

[82] Mooradian AD, Haas MJ, Batejko O, Hovsepyan M, Feman SS. Statins ameliorateendothelial barrier permeability changes in the cerebral tissue of streptozotocin-induced diabetic rats. Diabetes 2005;54:2977–82.

[83] Prasad R, Giri S, Nath N, Singh I, Singh AK. Inhibition of phosphoinositide 3kinase-Akt (protein kinase B)-nuclear factor-kappa B pathway by lovastatinlimits endothelial–monocyte cell interaction. J Neurochem 2005;94:204–14.

[84] Hempel A, Lindschau C, Maasch C, Mahn M, Bychkov R, Noll T, et al. Calciumantagonists ameliorate ischemia-induced endothelial cell permeability byinhibiting protein kinase C. Circulation 1999;99:2523–9.

[85] Abbruscato TJ, Davis TP. Combination of hypoxia/aglycemia compromises in vitroblood-brain barrier integrity. J Pharmacol Exp Ther 1999;289:668–75.

[86] Brown RC, Davis TP. Calciummodulation of adherens and tight junction function:a potential mechanism for blood-brain barrier disruption after stroke. Stroke2002;33:1706–11.

[87] Volpe M, Cosentino F. Abnormalities of endothelial function in the pathogenesisof stroke: the importance of endothelin. J Cardiovasc Pharmacol 2000;35:S45–48.

[88] McCarron RM, Chen Y, Tomori T, Strasser A, Mechoulam R, Shohami E, et al.Endothelial-mediated regulation of cerebral microcirculation. J Physiol Pharma-col 2006;57:133–44.

[89] Zinkel JL. Postischemic reperfusion: ultrastructural blood-brain barrier andhemodynamic correlative changes in an awake model of transient forebrainischemia. Neurosurgery Nov 2006;59(5):E1152.

[90] Lee JM, Zhai G, Liu Q, Gonzales ER, Yin K, Yan P, et al. Vascular permeabilityprecedes spontaneous intracerebral hemorrhage in stroke-prone spontaneouslyhypertensive rats. Stroke 2007;38:3289–91.

[91] Popescu BO. Still debating a cause and diagnostic criteria for Alzheimer's disease.J Cell Mol Med 2007;11:1225–6.

[92] Cedazo-Mínguez A. Apolipoprotein E and Alzheimer's disease: molecularmechanisms and therapeutic opportunities. J Cell Mol Med 2007;11:1227–38.

[93] OpricaM, Hjorth E, Spulber S, Popescu BO, AnkarcronaM,Winblad B, et al. Studieson brain volume, Alzheimer-related proteins and cytokines in mice with chronicoverexpression of IL-1 receptor antagonist. J Cell Mol Med 2007;11:810–25.

[94] Reid PC, Urano Y, Kodama T, Hamakubo T. Alzheimer's disease: cholesterol,membrane rafts, isoprenoids and statins. J Cell Mol Med 2007;11:383–92.

[95] Forero DA, Casadesus G, Perry G, Arboleda H. Synaptic dysfunction and oxidativestress in Alzheimer's disease: emerging mechanisms. J Cell Mol Med 2006;10:796–805.

[96] PopescuBO, Cedazo-MinguezA, Benedikz E, Nishimura T,Winblad B, AnkarcronaM,et al. Gamma-secretase activity of presenilin 1 regulates acetylcholine muscarinicreceptor-mediated signal transduction. J Biol Chem 2004;279:6455–64.

[97] Popescu BO, AnkarcronaM. Mechanisms of cell death in Alzheimer's disease: roleof presenilins. J Alzheimers Dis 2004;6:123–8.

[98] Desai BS, Monahan AJ, Carvey PM, Hendey B. Blood-brain barrier pathology inAlzheimer's and Parkinson's disease: implications for drug therapy. CellTransplant 2007;16(3):285–99.

[99] Deane R, Wu Z, Zlokovic BV. RAGE (yin) versus LRP (yang) balance regulatesAlzheimer amyloid beta-peptide clearance through transport across the blood-brain barrier. Stroke 2004;35:2628–31.

[100] Deane R, Du Yan S, Submamaryan RK, LaRue B, Jovanovic S, Hogg E, et al. RAGEmediates amyloid-beta peptide transport across the blood-brain barrier andaccumulation in brain. Nat Med 2003;9:907–13.

[101] Zlokovic BV. Clearing amyloid through the blood-brain barrier. J Neurochem2004;89:807–11.

[102] Deane R, Sagare A, Hamm K, Parisi M, LaRue B, Guo H, et al. IgG-assisted age-dependent clearance of Alzheimer's amyloid beta peptide by the blood-brainbarrier neonatal Fc receptor. J Neurosci 2005;25:11495–503.

[103] Cirrito JR, Deane R, Fagan AM, Spinner ML, Parsadanian M, Finn MB, et al. P-glycoprotein deficiency at the blood-brain barrier increases amyloid-β depositionin an Alzheimer disease mouse model. J Neurosci 2005;25:11495–503.

[104] Ujiie M, Dickstein DL, Carlow DA, Jefferies WA. Blood-brain barrier permeabilityprecedes senile plaque formation in an Alzheimer disease model. Microcircula-tion 2003;10:463–70.

[105] Banks WA, Terrell B, Farr SA, Robinson SM, Nonaka N, Morley JE. Passage ofamyloid beta protein antibody across the blood-brain barrier in amouse model ofAlzheimer's disease. Peptides 2002;23:2223–6.

[106] Methia N, André P, Hafezi-Moghadam A, Economopoulos M, Thomas KL, WagnerDD. ApoE deficiency compromises the blood brain barrier especially after injury.Mol Med 2001;7:810–5.

[107] Hafezi-Moghadam A, Thomas KL, Wagner DD. ApoE deficiency leads to aprogressive age-dependent blood-brain barrier leakage. Am J Physiol Cell Physiol2007;292:C1256–1262.

[108] Mulder M, Blokland A, van den Berg DJ, Schulten H, Bakker AH, Terwel D, et al.Apolipoprotein E protects against neuropathology induced by a high-fat diet andmaintains the integrity of the blood-brain barrier during aging. Lab Invest2001;81:953–60.

[109] Shibata M, Yamada S, Kumar SR, Calero M, Bading J, Frangione B, et al. Clearanceof Alzheimer's amyloid-ss(1–40) peptide from brain by LDL receptor-relatedprotein-1 at the blood-brain barrier. J Clin Invest 2000;106:1489–99.

[110] Algotsson A, Winblad B. The integrity of the blood-brain barrier in Alzheimer'sdisease. Acta Neurol Scand 2007;115:403–8.

105B.O. Popescu et al. / Journal of the Neurological Sciences 283 (2009) 99–106

Author's personal copy

[111] Bowman GL, Kaye JA, Moore M, Waichunas D, Carlson NE, Quinn JF. Blood-brainbarrier impairment in Alzheimer disease: stability and functional significance.Neurology 2007;68:1809–14.

[112] FormanMS, Lal D, Zhang B, Dabir DV, Swanson E, Lee VM, et al. Transgenic mousemodel of tau pathology in astrocytes leading to nervous system degeneration. JNeurosci Apr 6 2005;25(14):3539–50.

[113] Dickstein DL, Biron KE, Ujiie M, Pfeifer CG, Jeffries AR, Jefferies WA. Abeta peptideimmunization restores blood-brain barrier integrity in Alzheimer disease. FASEB JMar 2006;20(3):426–33.

[114] Schiera G, Proia P, Alberti C, Mineo M, Savettieri G, Di Liegro I. Neurons produceFGF2 and VEGF and secrete them at least in part by shedding extracellularvesicles. J Cell Mol Med 2007;11:1384–94.

[115] Lok J, Gupta P, Guo S, KimWJ, Whalen MJ, van Leyen K, et al. Cell–cell signaling inthe neurovascular unit. Neurochem Res 2007;32:2032–45.

[116] Rite I, Machado A, Cano J, Venero JL. Blood-brain barrier disruption induces in vivodegeneration of nigral dopaminergic neurons. J Neurochem 2007;101:1567–82.

[117] Whitton PS. Inflammation as a causative factor in the aetiology of Parkinson'sdisease. Br J Pharmacol 2007;150:963–76.

[118] Kortekaas R, Leenders KL, van Oostrom JC, Vaalburg W, Bart J, Willemsen AT, et al.Blood-brain barrier dysfunction in parkinsonian midbrain in vivo. Ann Neurol2005;57:176–9.

[119] Yokel RA. Blood-brain barrier flux of aluminum, manganese, iron and othermetals suspected to contribute to metal-induced neurodegeneration. J Alzhei-mers Dis 2006;10:223–53.

[120] Toescu EC. Normal brain ageing: models and mechanisms. Philos Trans R SocLond B Biol Sci 2005;360:2347–54.

106 B.O. Popescu et al. / Journal of the Neurological Sciences 283 (2009) 99–106