Open access

Platelets in Alzheimer’s Disease

Written By

Barbara Plagg and Christian Humpel

Submitted: 19 August 2014 Published: 18 November 2015

DOI: 10.5772/60535

From the Edited Volume

The Non-Thrombotic Role of Platelets in Health and Disease

Edited by Steve Kerrigan and Niamh Moran

Chapter metrics overview

1,740 Chapter Downloads

View Full Metrics

1. Introduction

Alzheimer’s disease (AD) is a progressive neurodegenerative disorder that gradually leads to severe cognitive impairment. The neuropathological hallmarks of AD include beta-amyloid plaques, tau neurofibrillary tangles, inflammation and glial responses, vascular dysfunction, synapse loss and cholinergic neurodegeneration. Currently, the diagnosis of possible or probable AD is based on a time consuming combination of clinical and psychological testing, imaging, and the analysis of three well-established biomarkers (beta-amyloid(42), total tau and phospho-tau-181) in cerebrospinal fluid. The search for biomarkers in blood is of high importance to avoid invasive lumbar puncture and to allow fast and easy analysis of a high number of patients. Biomarkers have been screened in blood of AD patients in plasma/serum, peripheral blood mononuclear cells (PBMCs), monocytes or also in platelets.

Platelets are interesting targets to study AD because they share some properties with neurons: they contain the neurotransmitter serotonin and the amyloid-precursor protein (APP), which produces the beta-amyloid, which aggregates in the brain of AD patients. Moreover, platelets are an easily accessible source of human cells. This review focuses on changes in the platelets of AD patients and will summarize (1) platelet activation including mean platelet volume, membrane fluidity and coated platelets, (2) serotonin metabolism, (3) APP isoforms and processing enzymes, including secretases, (4) oxidative stress and radicals, including nitric oxide metabolism and mitochondrial pathologies, including cytochrome-c oxidase and monoamino-oxidase-B and (5) enzymatic activity, such as glycogen synthase kinase-3 or phospholipase A2. In spite of all efforts, the discrepant results so far have prevented the establishment of a valid platelet-derived AD biomarker. With this survey we provide a detailed review about the major current findings on the potential use of platelet markers in AD.

Alzheimer’s disease (AD) is a progressive neurodegenerative disorder characterized by progressive deterioration in cognition leading to premature death. The neuropathological hallmarks of AD include beta-amyloid (Aβ) plaques, neurofibrillary tau tangles, inflammation and glial responses, synapse loss and cholinergic neurodegeneration among others. The worldwide prevalence of dementia was estimated as high as 24.2 million by 2005 and is expected to quadruplicate by 2050 [1, 2]. North America and Western Europe show the highest prevalence rate of dementia for persons >60 years [3]. Most dementia cases (70%) are attributed to AD, placing a considerable burden on society [2]. The increasing rate of AD cases not only stresses the establishment of efficient therapeutic intervention but also the identification of economic and reliable biomarkers. In both domains great efforts and smaller progress have been made in the last decades, while the desired breakthrough has yet to be awaited.

AD is classified into clinically indistinguishable early onset genetic AD (onset < 60 years) and late onset sporadic AD (> 60 years). Early onset AD accounts for approx. 1-5% of all cases and is associated with a more rapid progression of the neurodegenerative process and autosomal dominantly inherited mutations with a high penetrance. Sporadic AD accounts for >95% of all dementia cases and turned out to be a more heterogeneous disease. Currently, the diagnosis of possible or probable AD according to the NINCDS-ADRDA criteria is based on a time consuming combination of psychological testing, imaging, and the analysis of three well-established biomarkers (beta-amyloid(42), total tau and phospho-tau-181) in cerebrospinal fluid (CSF).

Biochemical markers mirror the physiological changes and can be objectively measured and evaluated as an indicator of pathological changes. A good biomarker must be sufficiently sensitive to detect early changes and specific to differentiate AD from clinically similar conditions. Besides high diagnostic and prognostic accuracy, biomarkers should allow global reproducibility with non-invasive and easy-to-perform tests. Within the search for peripheral AD-specific biomarkers, anucleated blood platelets have shown to be a promising target, since they represent the principal component of human blood affected by (early) biochemical alterations during AD. Moreover, platelets are an easily accessible source of human tissue and contain proteins found in neuronal cells.

In this chapter we aim to review changes in AD occurring in platelets to understand whether they may have a potential as putative biomarkers for diagnosing AD. We will focus on (1) platelet activation including mean platelet volume, membrane fluidity and coated platelets, (2) serotonin metabolism, (3) APP isoforms and processing enzymes, including secretases, (4) oxidative stress and radicals, including nitric oxide metabolism and mitochondrial pathologies, including cytochrome-c oxidase and monoamino-oxidase-B activity and (5) enzymatic activity, such as glycogen synthase kinase-3 or phospholipase A2. In spite of all efforts made, the discrepant results so far have prevented the establishment of a valid platelet-derived AD biomarker. With this survey we provide a detailed review about the major current findings on the potential use of platelet markers in AD.

Advertisement

2. Platelet activation

Vascular risk factors were traditionally considered to distinguish between vascular dementia and AD. However, in the last decade a series of studies revealed that vascular events are involved in the development of AD and the arbitrary classification into vascular dementia and AD is very much outdated [4-7]. Several studies reported vascular alterations such as an increased number of fragmented vessels, atrophic string vessels, changed vessel diameters, altered capillary membrane and collagen accumulation in the basement membrane of AD patients [8, 9]. Besides, deposits of Aβ in cerebral vessels (cerebral amyloid angiopathy (CAA)) induce severe damage of the vessel wall and alter the cerebral blood flow promoting the progression of AD [10]. Moreover, it became clear that AD associated alterations are not solely limited to the brain and occur in vessels and blood cells of the peripheral system [11].

Activation and aggregation of platelets are important steps for haemostasis at sites of vascular injury, while uncontrolled activation can trigger thrombotic vessel occlusion at sides of atherosclerotic plaque rupture [12] and lead to chronic inflammatory reactions. In AD, activated platelets are strongly linked to vascular processes and are proposed to be the missing link for the association between atherosclerotic events and AD [13]. Adhesion and activation of platelets on the vascular wall progressively lead to vascular inflammation and atherosclerosis, thus playing a key role in the development of AD-associated conditions. Increased platelet activation has been identified in the late 90s, due to damaged cerebral endothelial cells or membrane abnormalities in the AD brain [14].

Moreover, increased platelet activation, measured by GPIIb-IIIa complex activation or P-selectin expression is significantly higher in AD patients with fast cognitive decline compared to slow cognitive decline [13]. Furthermore it is known, that peripheral Aβ peptides contribute to platelet adhesion and activation in the initiation of thrombus formation [15, 16]. Recently it has been shown that activated platelets aggregate at sites of vascular Aβ promoting CAA by inducing platelet thrombus formation leading to vessel occlusion at vascular Aβ plaques [17, 18]. Because platelets are major players in blood flow alterations and vascular diseases, it was suggested that they do not only mirror AD related changes, but promote actively the progression of AD. In light of these findings it is conceivable that increased platelet activity could induce the progression of AD by contributing to peripheral vascular damage and endothelial senescence. Uncontrolled activation of platelets in AD-subjects may result in chronic inflammation mediating endothelial cell stress, which, in turn, may trigger platelet activation [19]. Alternatively, systemic inflammation in AD patients may result in platelet activation creating a vicious, self-amplifying circle [20]. Thus, it is conceivable that activated platelets contribute at different sites to the progression of AD.

References Effect on platelet activation
Sevush et al., 1998 in AD
Stellos et al., 2010, 2014 in AD with faster cognitive decline compared to AD with slow decline
in coronary artery disease with cognitive impairment

Table 1.

Platelet activation in AD

2.1. Effects on platelet volume

The platelet volume is a marker of platelet activation and thus involved in the pathophysiology of multiple pro-inflammatory diseases. Along with platelet activation, platelet volume values are considered indicators for vascular events linked to cerebral vascular dementia. In AD, both increased [21, 22] and decreased [23, 24] mean platelet volume has been reported. These heterogeneous results point out the importance of establishing methodological consensus in the isolation and processing of platelets, since they are very sensitive to cellular damage.

References Effect on platelet volume
Yagi et al., 1984 in vascular dementia compared to AD
Yesil et al., 2012 in AD
Wang et al., 2013 in AD and MCI; associated with MMSE
Liang et al., 2013 in vascular dementia and AD

Table 2.

Platelet volume in AD

2.2. Effects on membrane fluidity

Discrepant results have been published concerning platelet membrane fluidity: while decreased fluidity of cell membranes from platelets is associated with normal ageing, increased internal membrane fluidity is one of the first alterations to be reported in platelets [25, 26]. Increased platelet membrane fluidity becomes apparent by a decrease in the fluorescence anisotropy of 1,6-diphenyl-1,3,5-hexatriene (DPH) in labelled membranes. It was proposed, that this apparent abnormality of membranes in AD patients is due to an increase in internal membranes [26]. However, no significant differences in either cholesterol or phospholipid compared to healthy subjects has been observed [26], while an increase of intracellular platelet membranes in AD patients has been reported [27]. Other findings suggest furthermore that demented patients with increased platelet membrane fluidity display an earlier onset and a more rapid deterioration of cognitive symptoms compared to other demented subjects [25]. In light of these findings, platelet membrane fluidity was proposed as a biological risk factor for AD, since it is associated with significant contributions to the risk of developing AD [28, 29]. However, others failed to demonstrate differences in platelet membrane fluidity between AD patients and healthy controls, concluding that platelet membrane fluidity cannot be considered as antemortem biomarker for AD [30-32]. An increase in DPH fluorescence anisotropy has been reported [33-36] and as anisotropy is inversely related to membrane fluidity, these results indicate in contrast to the previously mentioned studies a decrease of the external and internal membrane fluidity in AD [33]. It was suggested that the short life span of platelets makes them less susceptible for long-term modifications. And finally, a reduced fluidity in the platelet inner mitochondrial membrane has been reported, which was associated with oxidative damage [37], while others correlated membrane fluidity with alterations of APP fragments [38].

References Effects on membrane fluidity
Zubenko et al., 1987, 1990 in familial AD by fluorescence anisotropy
Kozubski et al., 1999, 2002 in AD with 2 spin-labelled markers
Cohen et al., 1987: Piletz et al.,1991 by fluorescence anisotropy, ratio of cholesterol to phospholipid
Kaakkola et al., 1991 in AD and multi-infarct dementia compared to controls
Ortiz et al., 2008 fluidity of platelet inner mitochondrial membrane
Fernandes et al., 1999 unchanged in AD by fluorescence anisotropy
Kukull et al., 1992 unchanged
Kálmàn et al., 1994 in AD by fluorescence anisotropy
Cardoso et al., 2004 unchanged in AD compared to controls
Zainaghi et al., 2007 membrane fluidity correlated with alteration of APP fragments
Vignini et al., 2013 in AD by fluorescence anisotropy, worse profile for male patients
Hajimohammadreza et al., 1990 in AD by fluorescence anisotropy

Table 3.

Platelet membrane fluidity in AD

2.3. Coated platelets

Coated-platelets (PLTs) are a subset of platelets produced upon dual-agonist stimulation with collagen und thrombin retaining several procoagulant α-granule proteins on their surface [39]. PLTs are important for the coagulation cascade because of their ability to generate thrombin at sites of vascular damage. Effectively, it was suggested that increased levels of PLTs may be related to prothrombotic conditions [40], while decreased levels of PLTs were linked to an increased risk for haemorrhage [41]. It was also demonstrated that coated-platelet levels are elevated in amnestic mild cognitive impairment (MCI) and correlate with the progression of AD [40, 42-45]. Additionally, the same group showed that MCIs with elevated coated-platelet levels are more likely to develop AD, while they found no significant alterations in patients with frontotemporal lobe dementia [44, 45]. So far, coated platelets seem to be an interesting target, which needs to be reproduced internationally by other research groups.

Advertisement

3. Serotonin

Several studies report abnormalities in serotonin (5-HT) concentration as well as alterations in its uptake during AD, possibly linked to psychobehavioral problems such as e.g. depression [46-48]. Platelets accumulate high levels of serotonin in dense granules and release it upon activation [49]. Early studies failed to demonstrate altered serotonin processing in platelets [50, 51], while in the early 90s heterogeneous results regarding 5-HT Km (Michaelis-Menten constant) and maximal velocity (Vmax) binding affinity were published [52, 53, 54]. Compared to controls, patients with AD demonstrated in different studies significantly lower platelet serotonin concentrations [55-58]. However, an increased 5-HT concentration in low-density platelet populations was recently reported [59]. Likewise, heterogeneous results have been reported for serotonin uptake: both increased and decreased serotonin uptake was found in AD platelets [52, 60]. In light of these data and despite great efforts, so far platelet-derived serotonin has not been established as a reliable biomarker for AD.

References Effects on serotonin
Tukiainen et al., 1981 unchanged 5-HT uptake in
Andersson et al., 1991 unchanged maximum number of binding sites (Bmax) and binding affinity
Inestrosa et al., 1993 ability to accumulate 5-HT
Km and Vmax
Kumar et al., 1995a 5-HT concentration
Kumar et al., 1995b affinity of binding of 5-HT to the platelet membrane in AD (only females tested)
Arora et al., 1991 Vmax 5-HT-uptake in mild and moderate AD
Trend Vmax 5-HT-uptake in severe AD
Koren et al., 1993 [3H]-5-HT uptake
Mimica et al., 2008 5-HT concentrations in non-psychotic female and psychotic male AD patients compared to controls
Muck-Seler et al., 2009 5-HT concentrations in the late phase of AD compared to other phases and controls
Milovanovic et al., 2014 in low-density platelet populations
Prokselj et al., 2014 5-HT concentrations in AD compared to AD controls

Table 4.

Platelet 5-HT in AD

Vmax = maximum number of 5-HT uptake sites; Km = Michaelis-Menten constant; EOAD = Early Onset AD


Advertisement

4. Amyloid Precursor protein (APP) and secretases

4.1. APP isoforms and ratios

APP is an integral membrane protein with a large extracellular domain and a shorter, intracellular C-terminal tail. Three major APP isoforms (770, 751 and 695 kDa) have been described. APP 751 und APP 770 contain a Kunitz-type serine protease inhibitor domain (APP KPI), while APP 695 lacks this domain. The APP isoforms are cut by different enzymes (secretases) into smaller peptides, whereas sequential cleavage by β-secretase (BACE1) and γ-secretase (ADAM-10) generates the neurotoxic Aβ fragments. Conversely, cleavage by α-secretase precludes the formation of amyloid fragments by processing APP within the Aβ domain.

Platelets are of particular interest in AD research, because they contain high levels of APP [61-63]. In contrast to neuronal tissues where isoform 695 lacking the KPI domain is the most abundant one, platelets express mainly APP770 whereas APP695 is marginally present [64]. Platelets contain α-, β-, γ- secretase activities and generate different APP fragments: sAPPα, sAPPβ, the amyloidogenic fragment (C99) and Aβ peptides [65, 66]. Platelet APP is mainly processes by the α-secretase pathway releasing soluble APP (sAPP) [66] and predominantly Aβ(40). Both APP and Aβ are stored in α-granules of platelets and become released upon activation by agents like the physiological agonists thrombin and collagen. A recent study reports significant up-regulation of platelet APP isoforms compared to controls, and a correlation between APP mRNA levels and cognitive impairment [67]. The same group found significant up-regulation of platelet mRNA expression level of total APP and APP containing a KPI domain in patients with AD and frontotemporal lobe dementia compared to controls [68]. We have recently shown that platelet-secreted APPβ in MCI and AD is significantly increased when measured with ELISA compared to control subjects, while no changes in sAPPα are seen [69].

References APP expression and release
Vignini et al., 2013 mRNA of total APP and APP containing KPI domain in AD and FTLD
APP isoforms compared to controls
Marksteiner and Humpel, 2013 sAPP–β in MCI and AD compared to controls
unchanged sAPP-α in MCI and AD compared to controls

Table 5.

Platelet APP expression in AD

Several studies showed that the platelet APP ratio (defined as the ratio between the upper 130kDa and the lower 106-110kDa isoforms) is significantly lower in AD patients compared to controls and patients with other forms of dementia [70-77]. It seems that the alteration of platelet APP isoforms is an early event in AD and the ratio shows to be a consistent predictor for the conversion from MCI to AD. In fact, MCI subjects converting to AD showed significantly decreased APP ratios at baseline compared to other dementia forms and stable MCI subjects [78-80]. Furthermore, the APP ratio positively correlates with cognitive decline, i.e. the lower the ratio, the more severe the disease [38, 73]. Furthermore, it has been shown that carriers of the APOE4 allele are associated with a larger reduction in the APP ratio [81]. Moreover, administration of acetylcholine esterase inhibitors [82, 83] increases the ratio of APP forms in AD suggesting a possible effect of these drugs on APP trafficking in platelets. The proposed cut-off scores are around 0.56 with a sensitivity of 88% and specificity of 89% [80, 84]. Thus, the APP ratio has been proposed as a potential biomarker in prodromal AD-stages and a reliable indicator for the disease progress. Despite the promising homogeneous findings the validity of APP ratio as a useful supportive biomarker for AD diagnosis is not yet internationally established. Methodological problems including lack of sensitivity and diversity of the used antibodies and different isolation procedures of platelets may account for this problem.

References Effects on APP ratio
Di Luca et al., 1996, 1998 in AD and elderly patients with Down Syndrom; ratio correlated with the severity of the disease
Rosenberg et al., 1997 in AD compared to controls
Baskin et al., 2000, 2001 specific in AD compared to PD and HS (Hemorrhagic Stroke);
unchanged in cognitively normal young adults carrying PS-1 mutations
Padovani et al., 2001, 2002 in MCI and mild and very mild AD
Borroni et al., 2002, 2003, 2004 in MCIs converting to AD and early stages of probable AD
Liu et al., 2005 in AD; increases with galantamine treatment for 12 weeks
Tang et al., 2006 in AD
Zainaghi et al., 2007, 2012 in MCI converting to dementia upon follow-up
Srisawat et al., 2013 in Thai patients with AD

Table 6.

Platelet APP ratio in AD

4.2. Enzymatic activity: BACE1 and ADAM-10

APP is cleaved into secreted (soluble) APP (sAPP), smaller intracellular fragments and the Aβ peptides (40, 42 or 43 amino acids) by three secretases (α, β, γ). The α-secretase leads to the non-amyloidogenic pathway, while cleavage of APP by β -secretase (BACE1) and γ-secretase (ADAM-10) generates the toxic Aβ fragments. So far, several authors report increased BACE1 in AD by Western blotting or ELISA analysis [69, 73, 85, 86]. However, using a novel ELISA system a significant decrease of BACE1 N-terminal and C-terminal fragments has been reported in AD [87]. ADAM-10 is the major constitutive α-secretase for APP processing [88, 89]. To date, platelet ADAM10 (a disintegrin and metalloprotease) has been reported to be significantly reduced in platelets in AD [85, 90, 91] while others failed to detect changes in alpha- and beta-secretase activities in AD [92]. Moreover, reduction in ADAM-10 levels correlates with the progression and seems to be stage-dependent [91].

References Effects on secretases
Johnston et al., 2008 platelet membrane ß-secretase activity in AD compared to controls;
Colciaghi et al., 2002, 2004 ADAM10 in AD compared to controls
level of alpha APPs
BACE1 in AD
Tang et al., 2006 BACE1 in AD
ADAM10 in AD
Gorham et al., 2010 unchanged alpha- and beta-secretase activities and -ratio in AD and MCI
Decourt et al., 2013 BACE1 in AD (newly developed ELISA)
Manzine et al., 2013 ADAM10 in AD

Table 7.

Platelet secretase activity in AD

4.3. Activation of platelets by beta-amyloid

It is well known that different heterogeneous amyloidogenic peptides (Aβ(1-40), Aβ(1-42), Aβ(25-36)) as well as aggregated Aβ can induce platelet aggregation [15, 16]. Platelets store and release preferentially the 40 amino acid Aβ fragment in their granules upon stimulation with physiological agonists like thrombin, collagen or calcium ionophores [93, 61, 94, 95]. Once released, Aβ peptides trigger platelet activation, initiating a vicious feedback loop of platelet activation and Aβ release. Further, apoptotic stimuli significantly increase platelet Aβ(40) but not Aβ(42) suggesting that this pathway determines altered APP processing [96, 97]. Recently it was shown that Aβ induces platelet activation independent of known physiological agonists [16]. It was furthermore suggested that platelets modulate aggregation of soluble Aβ into fibrillar Aβ and facilitate platelet adhesion at vascular Aβ accumulations, contributing to the full occlusion of the affected vessel [18]. At this point it seems therefore likely that platelets and platelet-derived Aβ may contribute to a significant degree to the amyloid burden in the vascular walls promoting CAA in AD patients [96].

Advertisement

5. Oxidative stress, radicals and mitochondrial pathologies

Excessive chronic oxidative stress and production of radicals in the AD brain has been considered to promote cellular degeneration. Especially, nitric oxide (NO) and peroxynitrite (ONOO) are very reactive toxic radicals (ROS). It becomes more and more clear that a dysregulation of mitochondria and the involvement of cytochrome C-oxidase may play a role in this process. Vascular damage and endothelial dysfunction may play a role in AD and platelets serve as a source of oxidative stress. Mitochondria are the major sites responsible for more than 90 % of the ROS generation. In AD, mitochondrial DNA of cortical neurons was reported to induce excessive oxidative damage and increased DNA mutations [98-100]. Moreover, abnormal mitochondrial size and decreased mitochondrial number in AD and MCI are likely to increase ROS generation and oxidative damage [99, 101]. In platelets reduced Complex IV and Complex III activity has been repeatedly associated with AD [102-105, 32]. Subsequently, Aβ was shown to interact directly with mitochondria and inhibit platelet Complex IV activity inducing oxidative stress [106].

5.1. Nitric oxide and peroxynitrite

An increase in nitric oxide synthase (NOS) has been associated with normal aging and with AD [107, 108, 33]. Similarly to NO, peroxynitrite (ONOO) was found to be increased in AD patients and both NO and ONOO are linked to reduced Na+/K+-ATPase activity in platelet membranes of AD patients [108]. Additionally, carriers of the epsilon 4 allele of apolipoprotein E (APOE) show higher NOS compared to non-carriers [109]. In contrast, others found significantly lower NO concentrations in platelets of AD patients and a generally higher platelet aggregation rate. Since NO is known to inhibit platelet aggregation, NO might be responsible for the aggregation of platelets in the observed AD cohort [110].

References Effects on nitric oxide
Kawamoto et al., 2005 NO and peroxynitrite in AD
Vignini et al., 2007, 2013; NO and ONOO(-) peroxynitrite in AD
Marcourakis et al., 2008 NOS activity in APOE epsilon 4 carriers
Yu and Jia, 2009 NO and eNOS (endothelial nitric oxide synthase)

Table 8.

Platelet nitric oxide and peroxynitrite concentrations in AD

5.2. Cytochrome c oxidase

Cytochrome C oxidase (COX) is an enzyme located in mitochondria and may play a role in the production of radicals. In most studies, reduced platelet COX activity has been found in AD [111-113, 102, 103, 32, 104] but also in cognitively normal individuals with a maternal history of AD [114]. However, another study did not find any differences in COX activity in AD [115]. Recently, reduced mitochondrial COX activity has been found, which correlates to decreased mitochondrial membrane potential, resulting in higher lipid peroxides, superoxide radicals and protein carbonyls [111]. It has been proposed, that reduced COX activity causes higher tissue vulnerability and reduced oxygen availability [104].

References Effects on COX activity
Silva et al., 2013 COX, mitochondrial membrane potential , cytochrome c content
Burbaeva et al., 2012 COX in AD and MCI
Mosconi et al., 2011 COX activity in cognitively normal individuals with a history of maternal LOAD
Cardoso et al., 2004 15 % COX activity in AD despite COX subunits were present at normal levels
Bosetti et al., 2002 COX in AD and MCI
Van Zuylen et al., 1992 unchanged
Parker et al., 1990, 1991, 1994 COX in AD

Table 9.

Platelet COX activity in AD

5.3. Monoamino-oxidase B

Monoamino-oxidase-B (MAO-B) is an important enzyme located in the mitochondria and plays a role in metabolic processes of serotonin. Studies on platelet MAO-B activity yielded inconsistent results up to date: increased MAO-B activity was reported by several groups [116-126], while few researchers could not find any abnormalities [127,128]. The decline of mini-mental state examination scores (MMSE) preceded the elevation of MAO-B activity [116-119]. It has been suggested that MAO-B activity might be an indicator for severity and clinical progress in AD [57]. In another study, non-psychotic AD patients showed significantly higher platelet MAO-B activity, suggesting that MAO-B activity can differentiate between psychotic and non-psychotic subtypes of AD [56].

References Effects on MAO-B activity
Zellner et al., 2012 MAO-B activity correlated with MAO-B protein expression in AD
Mimica et al., 2008 in AD in non-psychotic patients
Soto et al., 1999 22% MAO-B activity in AD
Götz et al., 1998 MAO-B activity in AD
Bongioanni et al., 1996 MAO-B activity in AD
Ahlskop et al.,1996 unchanged
Konings et al., 1995 unchanged
Parnetti et al., 1992, 1994 MAO-B activity in LOAD compared to controls and EOAD
Regland et al., 1991 MAO-B activity in AD; after B12 therapy MAO-B activity was reduced to a normal level
Schneider et al., 1988 MAO-B activity in female AD without agitation and delusions
Danielczyk et al., 1988 MAO-B activity in AD (non-familial)
Alexopoulos et al., 1987 MAO-B activity in demented patients with and without depression
Adolfsson et al., 1980 MAO-B activity in AD

Table 10.

Platelet MAO-B activity in AD

Advertisement

6. Changes of other biological systems

6.1. GSK3β activity

Glycogen synthase kinase 3-beta (GSK3β) is involved in the regulation of glycogen synthesis by phosphorylating and inactivating the glycogen synthase and in the regulation of intracellular signalling pathways [129-131]. Moreover, in neural cell tissues GSK3β is the most important Tau kinase and has been linked to synaptic plasticity and neural injury [132, 133]. Therefore, deregulation of GSK3β has a significant impact on the formation of neurofibrillary tangles. GSK3β is also expressed in platelets and might be involved in platelet activation [134]. A substantial higher expression of GSK3β has been shown in AD and MCI platelets as compared to healthy controls, which also correlated with worse memory performance [135].

6.2. Phospholipase activity

It is known that AD is associated with chronic inflammatory responses and platelets express inflammatory mediators such as chemokines, interleukins, adhesive proteins and contain enzymes such as phospholipase-A2 (PLA2) [20]. Phospholipases are important platelet enzymes involved in the metabolism of membrane phospholipids and inflammatory synthesis. PLA2 and phospholipase C (PLC) are altered in peripheral blood cells and significantly decreased PLC activity in platelets of AD patients has been reported [136], suggesting aberrant phospholipase metabolism. Further, decreased PLA2 activity was found in human platelets [137, 138], which correlates with the degree of cognitive impairment and is modulated by cognitive training in healthy elders [139]. In contrast, an increased platelet PLA2 activity in individuals with AD has also been shown [140]. Thus, again the reports on these enzyme activities are very controversial and do not allow to establish a concluding interpretation.

References Effects on PLA2 and PLC activity
Krzystanek et al., 2007 PLA2 in AD
Gattaz et al., 1995, 1996, 2004 PLA2 in AD and MCI
Matushima et al., 1995, 1998 PLC in AD

Table 11.

Platelet PLA2 and PLC activity in AD

6.3. Overview of other platelet components found to be altered in AD

Several other biological systems have been studied in platelets of AD patients, such as e.g. decreased plasma antioxidant power levels [107], decreased cathepsin D [141], increased platelet glutamine synthetase-like protein level in MCI [112], increased NA,K-ATPase activities in AD [107], unchanged Vitamin E and cholesterol content between AD and controls [32] or increased phenolsulphotransferase activity in demented patients [142] or decreased platelet peripheral-type benzodiazepine binding [143]. However, we do not claim to provide a complete overview of all markers. Anyhow, none of these markers has been found to be a suitable reproducible marker for AD.

Advertisement

7. Platelets as biomarkers

Biomarkers must objectively reflect physiological processes linked to AD and should be globally reproducible with easy-to-perform tests [144]. Moreover, besides being sensitive enough to differentiate AD from clinically similar diseases, a good biomarker should be able to detect early disease-associated changes. In AD, abnormal metabolism of APP, hyperphosphorylation of Tau, induction of oxidative stress and inflammatory cascades result in pathological changes in liquid fluids. Currently, the laboratory diagnosis of AD is based on the combination of three CSF biomarkers yielding a sensitivity of >95% and specificity of >85% [145, 146]. However, considering the invasiveness of lumbar puncture and the growing incidence rate of AD, the need for biomarkers in more accessible body fluids is necessary. Blood measurements are minimally invasive and less time-consuming compared to CSF. The establishment of new markers in plasma/serum has proven to be difficult because changes mirror a broad spectrum of physiological processes not necessarily related to AD [144]. Moreover, changes in plasma/serum are very small and heterogeneous, thus making the search for reliable and sensitive biomarkers challenging.

Within the search for peripheral biomarkers in AD, blood platelets have been of great interest during the last decades. These anuclear blood cells share several homeostatic functions with neurons such as accumulation and release of neurotransmitters like serotonin, and expression of receptors and enzymes [147]. Moreover, human platelets have shown to be the most important source of more than 90% of the circulating APP [148, 149]. On grounds of these findings, platelets can be considered a valid peripheral model for the analysis of metabolic pathways linked to AD. Despite great efforts, to date no specific platelet biomarker has been successfully established, while some platelet components have shown to be of greater validity than others.

Overall, the so far published studies underline the need to establish concurrent methods in order to yield comparable results and avoid methodological diversity among institutes. Platelets contain several types of surface receptors responding to external stimuli with activation or inhibitory actions [150]. In response to physiological and non-physiological stimuli, platelet granules and their contents are released, thus requiring careful handling during isolation and experimentation in order to avoid activation. Effectively, different platelet isolation and processing procedures may account to a significant extent for the observed inter-rater differences between institutes. Moreover, different phases of the disease, comorbidities or medications need to be taken into consideration when evaluating platelet status in AD patients. Additionally, further analysis and classification of platelets with respect to their different features (e.g. density fraction) might be helpful to generate more significant data. Besides a homogeneous patient group, the selection of healthy, age-matched controls is a known challenge in AD-research and might additionally explain varying data. Therefore, the so far obtained results need to be replicated in large and homogeneous cohorts according to the same methodological protocol by independent researchers. To date, the use of platelet biomarkers in the diagnostic routine of AD can yet not be recommended and is in urgent need of final evaluation.

Advertisement

8. Conclusions

In conclusion, the search for platelet biomarker so far has highlighted some candidates worth further investigation, while the breakthrough in identifying one satisfyingly specific and sensitive marker has yet to be awaited. Overall, among the studied platelet components and according to the available data, the platelet APP ratio and COX activity seem the most promising candidates for establishing new peripheral biomarkers for AD.

Advertisement

Acknowledgments

This study has been supported by the Austrian Science Funds (P24734-B24).

References

  1. 1. Ferri CP, Prince M, Brayne C, Brodaty H, Fratiglioni L, Ganguli M, Hall K, Hasegawa K, Hendrie H, Huang Y, Jorm A, Mathers C, Menezes PR, Rimmer E, Scazufca M; Alzheimer's Disease International. Global prevalence of dementia: a Delphi consensus study. Lancet. 2005; 366(9503) 2112-2117.
  2. 2. Reitz C, Mayeux R. Alzheimer disease: epidemiology, diagnostic criteria, risk factors and biomarkers. Biochem Pharmacol. 2014; 88(4) 640-651.
  3. 3. Mayeux R, Stern Y. Epidemiology of Alzheimer disease. Cold Spring Harb Perspect Med. 2012; 2(8).
  4. 4. Schneider JA. High blood pressure and microinfarcts: a link between vascular risk factors, dementia, and clinical Alzheimer's disease. J Am Geriatr Soc. 2009; 57(11) 2146-2147.
  5. 5. Breteler MM. Vascular involvement in cognitive decline and dementia. Epidemiologic evidence from the Rotterdam Study and the Rotterdam Scan Study. Ann N Y Acad Sci. 2000; 903 457-465.
  6. 6. Purnell C, Gao S, Callahan CM, Hendrie HC. Cardiovascular risk factors and incident Alzheimer disease: a systematic review of the literature. Alzheimer Dis Assoc Disord. 2009; 23(1) 1-10.
  7. 7. Zlokovic BV. Neurovascular mechanisms of Alzheimer's neurodegeneration. Trends Neurosci. 2005; 28(4) 202-208.
  8. 8. Bailey TL, Rivara CB, Rocher AB, Hof PR. The nature and effects of cortical microvascular pathology in aging and Alzheimer's disease. Neurol Res. 2004; 26(5) 573-578.
  9. 9. Farkas E, Luiten PG. Cerebral microvascular pathology in aging and Alzheimer's disease. Prog Neurobiol. 2001; 64(6) 575-611.
  10. 10. Thal DR, Griffin WS, de Vos RA, Ghebremedhin E. Cerebral amyloid angiopathy and its relationship to Alzheimer's disease. Acta Neuropathol. 2008; 115(6) 599-609.
  11. 11. Hardy J. Does Abeta 42 have a function related to blood homeostasis? Neurochem Res. 2007; 32(4-5) 833-835.
  12. 12. Gawaz M, Langer H, May AE. Platelets in inflammation and atherogenesis. J Clin Invest. 2005; 115(12) 3378-3384.
  13. 13. Stellos K, Panagiota V, Kögel A, Leyhe T, Gawaz M, Laske C. Predictive value of platelet activation for the rate of cognitive decline in Alzheimer's disease patients. J Cereb Blood Flow Metab. 2010; 30(11) 1817-1820.
  14. 14. Sevush S, Jy W, Horstman LL, Mao WW, Kolodny L, Ahn YS. Platelet activation in Alzheimer disease. Arch Neurol. 1998; 55(4) 530-536.
  15. 15. Canobbio I, Catricalà S, Di Pasqua LG, Guidetti G, Consonni A, Manganaro D, Torti M. Immobilized amyloid Aβ peptides support platelet adhesion and activation. FEBS Lett. 2013; 587(16) 2606-2611.
  16. 16. Sonkar VK, Kulkarni PP, Dash D. Amyloid β peptide stimulates platelet activation through RhoA-dependent modulation of actomyosin organization. FASEB J. 2014; 28(4) 1819-1829.
  17. 17. Roher AE, Esh CL, Kokjohn TA, Castaño EM, Van Vickle GD, Kalback WM, Patton RL, Luehrs DC, Daugs ID, Kuo YM, Emmerling MR, Soares H, Quinn JF, Kaye J, Connor DJ, Silverberg NB, Adler CH, Seward JD, Beach TG, Sabbagh MN. Amyloid beta peptides in human plasma and tissues and their significance for Alzheimer's disease. Alzheimers Dement. 2009; 5(1) 18-29.
  18. 18. Gowert NS, Donner L, Chatterjee M, Eisele YS, Towhid ST, Münzer P, Walker B, Ogorek I, Borst O, Grandoch M, Schaller M, Fischer JW, Gawaz M, Weggen S, Lang F, Jucker M, Elvers M. Blood platelets in the progression of Alzheimer's disease. PLoS One. 2014; 9(2).
  19. 19. Catricala S, Torti M, Ricevuti G. Alzheimer disease and platelets: how's that relevant. Immun Ageing. 2012 Sep 17;9(1):20. Casoli T, Di Stefano G, Balietti M, Solazzi M, Giorgetti B, Fattoretti P. Peripheral inflammatory biomarkers of Alzheimer's disease: the role of platelets. Biogerontology. 2010; 11(5) 627-633.
  20. 20. Casoli T, Di Stefano G, Balietti M, Solazzi M, Giorgetti B, Fattoretti P. Peripheral inflammatory biomarkers of Alzheimer's disease: the role of platelets. Biogerontology. 2010; 11(5) 627-633.
  21. 21. Yagi T, Kohashi N, Katori R, Miyazaki M. [Mean platelet volume, platelet count and platelet retention rate in cerebral vascular disease and Alzheimer type dementia]. Nihon Ronen Igakkai Zasshi. 1984; 21(6) 545-549.
  22. 22. Yesil Y, Kuyumcu ME, Cankurtaran M, Uz B, Kara A, Kilic MK, Halil M, Ulger Z, Yavuz BB, Haznedaroğlu IC, Ariogul S. Increased mean platelet volume (MPV) indicating the vascular risk in Alzheimer's disease (AD). Arch Gerontol Geriatr. 2012; 55(2) 257-260.
  23. 23. Wang RT, Jin D, Li Y, Liang QC. Decreased mean platelet volume and platelet distribution width are associated with mild cognitive impairment and Alzheimer's disease. J Psychiatr Res. 2013; 47(5) 644-649.
  24. 24. Liang QC, Jin D, Li Y, Wang RT. Mean platelet volume and platelet distribution width in vascular dementia and Alzheimer's disease. Platelets. 2013, 1-6.
  25. 25. Zubenko GS, Cohen BM, Reynolds CF 3rd, Boller F, Teply I, Chojnacki B. Platelet membrane fluidity identifies a clinical subtype of Alzheimer's disease. Prog Neuropsychopharmacol Biol Psychiatry. 1987;11(6) 683-699.
  26. 26. Cohen BM, Zubenko GS, Babb SM. Abnormal platelet membrane composition in Alzheimer's-type dementia. Life Sci. 1987; 40(25) 2445-2451.
  27. 27. Piletz JE, Sarasua M, Whitehouse P, Chotani M. Intracellular membranes are more fluid in platelets of Alzheimer's disease patients. Neurobiol Aging. 1991; 12(5) 401-406.
  28. 28. Zubenko GS, Kopp U, Seto T, Firestone LL. Platelet membrane fluidity individuals at risk for Alzheimer's disease: a comparison of results from fluorescence spectroscopy and electron spin resonance spectroscopy. Psychopharmacology (Berl). 1999; 145(2) 175-180.
  29. 29. Kozubski W, Swiderek M, Kloszewska I, Gwozdzinski K, Watala C. Blood platelet membrane fluidity and the exposition of membrane protein receptors in Alzheimer disease (AD) patients--preliminary Study. Alzheimer Dis Assoc Disord. 2002; 16(1) 52-54.
  30. 30. Kukull WA, Hinds TR, Schellenberg GD, van Belle G, Larson EB. Increased platelet membrane fluidity as a diagnostic marker for Alzheimer's disease: a test in population-based cases and controls. Neurology. 1992; 42(3 Pt 1) 607-614.
  31. 31. Fernandes MA, Proença MT, Nogueira AJ, Oliveira LM, Santiago B, Santana I, Oliveira CR. Effects of apolipoprotein E genotype on blood lipid composition and membrane platelet fluidity in Alzheimer's disease. Biochim Biophys Acta. 1999; 1454(1) 89-96.
  32. 32. Cardoso SM, Proença MT, Santos S, Santana I, Oliveira CR. Cytochrome c oxidase is decreased in Alzheimer's disease platelets. Neurobiol Aging. 2004; 25(1) 105-110.
  33. 33. Vignini A, Giusti L, Raffaelli F, Giulietti A, Salvolini E, Luzzi S, Provinciali L, Mazzanti L, Nanetti L. Impact of gender on platelet membrane functions of Alzheimer's disease patients. Exp Gerontol. 2013; 48(3) 319-325.
  34. 34. Kaakkola S, Rosenberg PH, Alila A, Erkinjuntti T, Sulkava R, Palo J. Platelet membrane fluidity in Alzheimer's disease and multi-infarct dementia: a spin label study. Acta Neurol Scand. 1991; 84(1) 18-21.
  35. 35. Kálmán J, Dey I, Ilona SV, Matkovics B, Brown D, Janka Z, Farkas T, Joó F. Platelet membrane fluidity and plasma malondialdehyde levels in Alzheimer's demented patients with and without family history of dementia. Biol Psychiatry. 1994; 35(3) 190-194.
  36. 36. Hajimohammadreza I, Brammer MJ, Eagger S, Burns A, Levy R. Platelet and erythrocyte membrane changes in Alzheimer's disease. Biochim Biophys Acta. 1990; 1025(2) 208-214.
  37. 37. Ortiz GG, Pacheco-Moisés F, El Hafidi M, Jiménez-Delgado A, Macías-Islas MA, Rosales Corral SA, de la Rosa AC, Sánchez-González VJ, Arias-Merino ED, Velázquez-Brizuela IE. Detection of membrane fluidity in submitochondrial particles of platelets and erythrocyte membranes from Mexican patients with Alzheimer disease by intramolecular excimer formation of 1,3 dipyrenylpropane. Dis Markers. 2008; 24(3) 151-156.
  38. 38. Zainaghi IA, Forlenza OV, Gattaz WF. Abnormal APP processing in platelets of patients with Alzheimer's disease: correlations with membrane fluidity and cognitive decline. Psychopharmacology (Berl). 2007; 192(4) 547-553.
  39. 39. Alberio L, Safa O, Clemetson KJ, Esmon CT, Dale GL. Surface expression and functional characterization of alpha-granule factor V in human platelets: effects of ionophore A23187, thrombin, collagen, and convulxin. Blood. 2000; 95(5) 1694-1702.
  40. 40. Prodan CI, Joseph PM, Vincent AS, Dale GL. Coated-platelets in ischemic stroke: differences between lacunar and cortical stroke. J Thromb Haemost. 2008; 6(4) 609-614.
  41. 41. Prodan CI, Vincent AS, Padmanabhan R, Dale GL. Coated-platelet levels are low in patients with spontaneous intracerebral hemorrhage. Stroke. 2009; 40(7) 2578-2580.
  42. 42. Prodan CI, Szasz R, Vincent AS, Ross ED, Dale GL. Coated-platelets retain amyloid precursor protein on their surface. Platelets. 2006; 17(1) 56-60.
  43. 43. Prodan CI, Ross ED, Vincent AS, Dale GL. Coated-platelets are higher in amnestic versus nonamnestic patients with mild cognitive impairment. Alzheimer Dis Assoc Disord. 2007; 21(3) 259-261.
  44. 44. Prodan CI, Ross ED, Vincent AS, Dale GL. Differences in coated-platelet production between frontotemporal dementia and Alzheimer disease. Alzheimer Dis Assoc Disord. 2009; 23(3) 234-237.
  45. 45. Prodan CI, Ross ED, Stoner JA, Cowan LD, Vincent AS, Dale GL. Coated-platelet levels and progression from mild cognitive impairment to Alzheimer disease. Neurology. 2011; 76(3) 247-52.
  46. 46. Assal F, Alarcón M, Solomon EC, Masterman D, Geschwind DH, Cummings JL. Association of the serotonin transporter and receptor gene polymorphisms in neuropsychiatric symptoms in Alzheimer disease. Arch Neurol. 2004; 61(8) 1249-1253.
  47. 47. Curcio CA, Kemper T. Nucleus raphe dorsalis in dementia of the Alzheimer type: neurofibrillary changes and neuronal packing density. J Neuropathol Exp Neurol. 1984; 43(4) 359-368.
  48. 48. Zubenko GS, Moossy J, Martinez AJ, Rao G, Claassen D, Rosen J, Kopp U. Neuropathologic and neurochemical correlates of psychosis in primary dementia. Arch Neurol. 1991; 48(6) 619-624.
  49. 49. De Clerck F, Xhonneux B, Leysen J, Janssen PA. Evidence for functional 5-HT2 receptor sites on human blood platelets. Biochem Pharmacol. 1984; 33(17) 2807-2811.
  50. 50. Tukiainen E, Wikström J, Kilpeläinen H. Uptake of 5-hydroxytryptamine by blood platelets in Huntington's chorea and Alzheimer type of presenile dementia. Med Biol. 1981; 59(2) 116-120.
  51. 51. Andersson A, Adolfsson R, Eriksson K, Marcusson J. Platelet [3H]paroxetine binding to 5-HT uptake sites in Alzheimer's disease. Neurobiol Aging. 1991; 12(5) 531-534.
  52. 52. Arora RC, Emery OB, Meltzer HY. Serotonin uptake in the blood platelets of Alzheimer's disease patients. Neurology. 1991; 41(8) 1307-1309.
  53. 53. Inestrosa NC, Alarcón R, Arriagada J, Donoso A, Alvarez J. Platelet of Alzheimer patients: increased counts and subnormal uptake and accumulation of [14C]5-hydroxytryptamine. Neurosci Lett. 1993; 163(1) 8-10.
  54. 54. Kumar AM, Kumar M, Sevush S, Ruiz J, Eisdorfer C. Serotonin uptake and its kinetics in platelets of women with Alzheimer's disease. Psychiatry Res. 1995; 59(1-2) 145-150.
  55. 55. Prokšelj T, Jerin A, Muck-Seler D, Kogoj A. Decreased platelet serotonin concentration in Alzheimer's disease with involuntary emotional expression disorder. Neurosci Lett. 2014; 578 71-74.
  56. 56. Mimica N, Mück-Seler D, Pivac N, Mustapić M, Dezeljin M, Stipcević T, Presecki P, Radonić E, Folnegović-Smalc V. Platelet serotonin and monoamine oxidase in Alzheimer's disease with psychotic features. Coll Antropol. 2008; 32 Suppl 1 119-122.
  57. 57. Muck-Seler D, Presecki P, Mimica N, Mustapic M, Pivac N, Babic A, Nedic G, Folnegovic-Smalc V. Platelet serotonin concentration and monoamine oxidase type B activity in female patients in early, middle and late phase of Alzheimer's disease. Prog Neuropsychopharmacol Biol Psychiatry. 2009; 33(7) 1226-1231.
  58. 58. Kumar AM, Sevush S, Kumar M, Ruiz J, Eisdorfer C. Peripheral serotonin in Alzheimer's disease. Neuropsychobiology. 1995; 32(1) 9-12.
  59. 59. Milovanovic M, Eriksson K, Winblad B, Nilsson S, Lindahl TL, Post C, Järemo P. Alzheimer and platelets: Low-density platelet populations reveal increased serotonin content in Alzheimer type dementia. Clin Biochem. 2014.
  60. 60. Koren P, Diver-Haber A, Adunsky A, Rabinowitz M, Hershkowitz M. Uptake of serotonin into platelets of senile dementia of the Alzheimer type patients. J Gerontol. 1993; 48(3) B93-96.
  61. 61. Bush AI, Martins RN, Rumble B, Moir R, Fuller S, Milward E, Currie J, Ames D, Weidemann A, Fischer P, et al. The amyloid precursor protein of Alzheimer's disease is released by human platelets. J Biol Chem. 1990; 265(26) 15977-15983.
  62. 62. Gardella JE, Ghiso J, Gorgone GA, Marratta D, Kaplan AP, Frangione B, Gorevic PD. Intact Alzheimer amyloid precursor protein (APP) is present in platelet membranes and is encoded by platelet mRNA. Biochem Biophys Res Commun. 1990; 173(3) 1292-1298.
  63. 63. Di Luca M, Colciaghi F, Pastorino L, Borroni B, Padovani A, Cattabeni F. Platelets as a peripheral district where to study pathogenetic mechanisms of alzheimer disease: the case of amyloid precursor protein. Eur J Pharmacol. 2000; 405(1-3) 277-283.
  64. 64. Evin G, Li QX. Platelets and Alzheimer's disease: Potential of APP as a biomarker. World J Psychiatry. 2012; 2(6) 102-113.
  65. 65. Li QX, Whyte S, Tanner JE, Evin G, Beyreuther K, Masters CL. Secretion of Alzheimer's disease Abeta amyloid peptide by activated human platelets. Lab Invest. 1998; 78(4) 461-469.
  66. 66. Li QX, Cappai R, Evin G, Tanner JE, Gray CW, Beyreuther K, Masters CL. Products of the Alzheimer's disease amyloid precursor protein generated by b-secretase are present in human platelets, and secreted upon degranulation. Am J Alzheimers Dis. 1998; 13 236–244.
  67. 67. Vignini A, Sartini D, Morganti S, Nanetti L, Luzzi S, Provinciali L, Mazzanti L, Emanuelli M. Platelet amyloid precursor protein isoform expression in Alzheimer's disease: evidence for peripheral marker. Int J Immunopathol Pharmacol. 2011; 24(2) 529-534.
  68. 68. Vignini A, Morganti S, Salvolini E, Sartini D, Luzzi S, Fiorini R, Provinciali L, Di Primio R, Mazzanti L, Emanuelli M. Amyloid precursor protein expression is enhanced in human platelets from subjects with Alzheimer's disease and frontotemporal lobar degeneration: a real-time PCR study. Exp Gerontol. 2013; 48(12) 1505-1508.
  69. 69. Marksteiner J, Humpel C. Platelet-derived secreted amyloid-precursor protein-β as a marker for diagnosing Alzheimer's disease. Curr Neurovasc Res. 2013; 10(4) 297-303.
  70. 70. Di Luca M, Pastorino L, Cattabeni F, Zanardi R, Scarone S, Racagni G, Smeraldi E, Perez J. Abnormal pattern of platelet APP isoforms in Alzheimer disease and Down syndrome. Arch Neurol. 1996; 53(11) 1162-1166.
  71. 71. Di Luca M, Pastorino L, Bianchetti A, Perez J, Vignolo LA, Lenzi GL, Trabucchi M, Cattabeni F, Padovani A. Differential level of platelet amyloid beta precursor protein isoforms: an early marker for Alzheimer disease. Arch Neurol. 1998; 55(9) 1195-1200.
  72. 72. Borroni B, Colciaghi F, Archetti S, Marcello E, Caimi L, Di Luca M, Padovani A. Predicting cognitive decline in Alzheimer disease. Role of platelet amyloid precursor protein. Alzheimer Dis Assoc Disord. 2004; 18(1) 32-34.
  73. 73. Tang K, Hynan LS, Baskin F, Rosenberg RN. Platelet amyloid precursor protein processing: a bio-marker for Alzheimer's disease. J Neurol Sci. 2006; 240(1-2) 53-58.
  74. 74. Zainaghi IA, Talib LL, Diniz BS, Gattaz WF, Forlenza OV. Reduced platelet amyloid precursor protein ratio (APP ratio) predicts conversion from mild cognitive impairment to Alzheimer's disease. J Neural Transm. 2012; 119(7) 815-819.
  75. 75. Baskin F, Rosenberg RN, Iyer L, Hynan L, Cullum CM. Platelet APP isoform ratios correlate with declining cognition in AD. Neurology. 2000; 54(10) 1907-9.
  76. 76. Baskin F, Rosenberg RN, Iyer L, Schellenberg GD, Hynan L, Nee LE. Platelet APP isoform ratios in asymptomatic young adults expressing an AD-related presenilin-1 mutation. J Neurol Sci. 2001; 183(1) 85-88.
  77. 77. Srisawat C, Junnu S, Peerapittayamongkol C, Futrakul A, Soi-ampornkul R, Senanarong V, Praditsuwan R, Assantachai P, Neungton N. The platelet amyloid precursor protein ratio as a diagnostic marker for Alzheimer's disease in Thai patients. J Clin Neurosci. 2013; 20(5) 644-8.
  78. 78. Borroni B, Colciaghi F, Corsini P, Akkawi N, Rozzini L, Del Zotto E, Talarico G, Cattabeni F, Lenzi GL, Di Luca M, Padovani A. Early stages of probable Alzheimer disease are associated with changes in platelet amyloid precursor protein forms. Neurol Sci. 2002; 23(5) 207-210.
  79. 79. Padovani A, Borroni B, Colciaghi F, Pettenati C, Cottini E, Agosti C, Lenzi GL, Caltagirone C, Trabucchi M, Cattabeni F, Di Luca M. Abnormalities in the pattern of platelet amyloid precursor protein forms in patients with mild cognitive impairment and Alzheimer disease. Arch Neurol. 2002; 59(1) 71-75.
  80. 80. Borroni B, Colciaghi F, Caltagirone C, Rozzini L, Broglio L, Cattabeni F, Di Luca M, Padovani A. Platelet amyloid precursor protein abnormalities in mild cognitive impairment predict conversion to dementia of Alzheimer type: a 2-year follow-up study. Arch Neurol. 2003; 60(12) 1740-1744.
  81. 81. Rosenberg RN, Baskin F, Fosmire JA, Risser R, Adams P, Svetlik D, Honig LS, Cullum CM, Weiner MF. Altered amyloid protein processing in platelets of patients with Alzheimer disease. Arch Neurol. 1997; 54(2) 139-144.
  82. 82. Borroni B, Colciaghi F, Pastorino L, Pettenati C, Cottini E, Rozzini L, Monastero R, Lenzi GL, Cattabeni F, Di Luca M, Padovani A. Amyloid precursor protein in platelets of patients with Alzheimer disease: effect of acetylcholinesterase inhibitor treatment. Arch Neurol. 2001; 58(3) 442-446.
  83. 83. Liu HC, Chi CW, Ko SY, Wang HC, Hong CJ, Lin KN, Wang PN, Liu TY. Cholinesterase inhibitor affects the amyloid precursor protein isoforms in patients with Alzheimer's disease. Dement Geriatr Cogn Disord. 2005; 19(5-6) 345-8.
  84. 84. Padovani A, Pastorino L, Borroni B, Colciaghi F, Rozzini L, Monastero R, Perez J, Pettenati C, Mussi M, Parrinello G, Cottini E, Lenzi GL, Trabucchi M, Cattabeni F, Di Luca M. Amyloid precursor protein in platelets: a peripheral marker for the diagnosis of sporadic AD. Neurology. 2001; 57(12) 2243-8.
  85. 85. Colciaghi F, Marcello E, Borroni B, Zimmermann M, Caltagirone C, Cattabeni F, Padovani A, Di Luca M. Platelet APP, ADAM 10 and BACE alterations in the early stages of Alzheimer disease. Neurology. 2004; 62(3) 498-501.
  86. 86. Johnston JA, Liu WW, Coulson DT, Todd S, Murphy S, Brennan S, Foy CJ, Craig D, Irvine GB, Passmore AP. Platelet beta-secretase activity is increased in Alzheimer's disease. Neurobiol Aging. 2008; 29(5) 661-668.
  87. 87. Decourt B, Walker A, Gonzales A, Malek-Ahmadi M, Liesback C, Davis KJ, Belden CM, Jacobson SA, Sabbagh MN. Can platelet BACE1 levels be used as a biomarker for Alzheimer's disease? Proof-of-concept study. Platelets. 2013;24(3):235-8. Marksteiner J, Humpel C. Platelet-derived secreted amyloid-precursor protein-β as a marker for diagnosing Alzheimer's disease. Curr Neurovasc Res. 2013; 10(4) 297-303.
  88. 88. Kuhn PH, Koroniak K, Hogl S, Colombo A, Zeitschel U, Willem M, Volbracht C, Schepers U, Imhof A, Hoffmeister A, Haass C, Roßner S, Bräse S, Lichtenthaler SF. Secretome protein enrichment identifies physiological BACE1 protease substrates in neurons. EMBO J. 2012; 31(14) 3157-68.
  89. 89. Jorissen E, Prox J, Bernreuther C, Weber S, Schwanbeck R, Serneels L, Snellinx A, Craessaerts K, Thathiah A, Tesseur I, Bartsch U, Weskamp G, Blobel CP, Glatzel M, De Strooper B, Saftig P. The disintegrin/metalloproteinase ADAM10 is essential for the establishment of the brain cortex. J Neurosci. 2010; 30(14) 4833-44.
  90. 90. Colciaghi F, Borroni B, Pastorino L, Marcello E, Zimmermann M, Cattabeni F, Padovani A, Di Luca M. [alpha]-Secretase ADAM10 as well as [alpha]APPs is reduced in platelets and CSF of Alzheimer disease patients. Mol Med. 2002; 8(2) 67-74.
  91. 91. Manzine PR, de França Bram JM, Barham EJ, do Vale Fde A, Selistre-de-Araújo HS, Cominetti MR, Iost Pavarini SC. ADAM10 as a biomarker for Alzheimer's disease: a study with Brazilian elderly. Dement Geriatr Cogn Disord. 2013; 35(1-2) 58-66.
  92. 92. Gorham P, Bark N, Björkhem I, Meaney S, Crisby M. Platelet alpha- and beta- secretase activities are not significantly affected by dementia or mild cognitive impairment in Swedish patients. Curr Alzheimer Res. 2010; 7(2) 134-139.
  93. 93. Li QX, Fuller SJ, Beyreuther K, Masters CL. The amyloid precursor protein of Alzheimer disease in human brain and blood. J Leukoc Biol. 1999; 66(4) 567-574.
  94. 94. Kokjohn TA, Van Vickle GD, Maarouf CL, Kalback WM, Hunter JM, Daugs ID, Luehrs DC, Lopez J, Brune D, Sue LI, Beach TG, Castaño EM, Roher AE. Chemical characterization of pro-inflammatory amyloid-beta peptides in human atherosclerotic lesions and platelets. Biochim Biophys Acta. 2011; 1812(11) 1508-1514.
  95. 95. Skovronsky DM, Lee VM, Praticò D. Amyloid precursor protein and amyloid beta peptide in human platelets. Role of cyclooxygenase and protein kinase C. J Biol Chem. 2001; 276(20) 17036-17043.
  96. 96. Casoli T, Di Stefano G, Giorgetti B, Grossi Y, Balietti M, Fattoretti P, Bertoni-Freddari C. Release of beta-amyloid from high-density platelets: implications for Alzheimer's disease pathology. Ann N Y Acad Sci. 2007; 1096 170-178.
  97. 97. Casoli T, Di Stefano G, Giorgetti B, Balietti M, Recchioni R, Moroni F, Marcheselli F, Bernardini G, Fattoretti P, Bertoni-Freddari C. Platelet as a physiological model to investigate apoptotic mechanisms in Alzheimer beta-amyloid peptide production. Mech Ageing Dev. 2008; 129(3) 154-162.
  98. 98. Corral-Debrinski M, Horton T, Lott MT, Shoffner JM, McKee AC, Beal MF, Graham BH, Wallace DC. Marked changes in mitochondrial DNA deletion levels in Alzheimer brains. Genomics. 1994; 23(2) 471-476.
  99. 99. Hirai K, Aliev G, Nunomura A, Fujioka H, Russell RL, Atwood CS, Johnson AB, Kress Y, Vinters HV, Tabaton M, Shimohama S, Cash AD, Siedlak SL, Harris PL, Jones PK, Petersen RB, Perry G, Smith MA. Mitochondrial abnormalities in Alzheimer's disease. J Neurosci. 2001; 21(9) 3017-3023.
  100. 100. Yan MH, Wang X, Zhu X. Mitochondrial defects and oxidative stress in Alzheimer disease and Parkinson disease. Free Radic Biol Med. 2013; 62 90-101.
  101. 101. Wang X, Su B, Lee HG, Li X, Perry G, Smith MA, Zhu X. Impaired balance of mitochondrial fission and fusion in Alzheimer's disease. J Neurosci. 2009; 29(28) 9090-9103.
  102. 102. Parker WD Jr, Filley CM, Parks JK. Cytochrome oxidase deficiency in Alzheimer's disease. Neurology. 1990; 40(8) 1302-1303.
  103. 103. Parker WD Jr, Mahr NJ, Filley CM, Parks JK, Hughes D, Young DA, Cullum CM. Reduced platelet cytochrome c oxidase activity in Alzheimer's disease. Neurology. 1994; 44(6) 1086-1090.
  104. 104. Bosetti F, Brizzi F, Barogi S, Mancuso M, Siciliano G, Tendi EA, Murri L, Rapoport SI, Solaini G. Cytochrome c oxidase and mitochondrial F1F0-ATPase (ATP synthase) activities in platelets and brain from patients with Alzheimer's disease. Neurobiol Aging. 2002; 23(3) 371-376.
  105. 105. Valla J, Schneider L, Niedzielko T, Coon KD, Caselli R, Sabbagh MN, Ahern GL, Baxter L, Alexander G, Walker DG, Reiman EM. Impaired platelet mitochondrial activity in Alzheimer's disease and mild cognitive impairment. Mitochondrion. 2006; 6(6) 323-330.
  106. 106. Crouch PJ, Blake R, Duce JA, Ciccotosto GD, Li QX, Barnham KJ, Curtain CC, Cherny RA, Cappai R, Dyrks T, Masters CL, Trounce IA. Copper-dependent inhibition of human cytochrome c oxidase by a dimeric conformer of amyloid-beta1-42. J Neurosci. 2005; 25(3) 672-9.
  107. 107. Kawamoto EM, Munhoz CD, Glezer I, Bahia VS, Caramelli P, Nitrini R, Gorjão R, Curi R, Scavone C, Marcourakis T. Oxidative state in platelets and erythrocytes in aging and Alzheimer's disease. Neurobiol Aging. 2005; 26(6) 857-864.
  108. 108. Vignini A, Nanetti L, Moroni C, Tanase L, Bartolini M, Luzzi S, Provinciali L, Mazzanti L. Modifications of platelet from Alzheimer disease patients: a possible relation between membrane properties and NO metabolites. Neurobiol Aging. 2007; 28(7) 987-994.
  109. 109. Marcourakis T, Bahia VS, Kawamoto EM, Munhoz CD, Gorjão R, Artes R, Kok F, Caramelli P, Nitrini R, Curi R, Scavone C. Apolipoprotein E genotype is related to nitric oxide production in platelets. Cell Biochem Funct. 2008; 26(8) 852-858.
  110. 110. Yu J, Jia JP. [Platelet function in patients with Alzheimer disease: analysis of 40 cases]. Zhonghua Yi Xue Za Zhi. 2009; 89(3) 183-186.
  111. 111. Silva DF, Santana I, Esteves AR, Baldeiras I, Arduino DM, Oliveira CR, Cardoso SM. Prodromal metabolic phenotype in MCI cybrids: implications for Alzheimer's disease. Curr Alzheimer Res. 2013; 10(2) 180-190.
  112. 112. Burbaeva GSh, Boksha IS, Savushkina OK, Turishcheva MS, Tereshkina EB, Starodubtseva LI, Gavrilova SI, Fedorova IaB, Zhuravin IA. [Platelet cytochrome c-oxidase and glutamine synthetase-like protein in patients with mild cognitive impairment]. Zh Nevrol Psikhiatr Im S S Korsakova. 2012; 112(2) 55-58.
  113. 113. Parker WD Jr. Cytochrome oxidase deficiency in Alzheimer's disease. Ann N Y Acad Sci. 1991; 640 59-64.
  114. 114. Mosconi L, de Leon M, Murray J, E L, Lu J, Javier E, McHugh P, Swerdlow RH. Reduced mitochondria cytochrome oxidase activity in adult children of mothers with Alzheimer's disease. J Alzheimers Dis. 2011; 27(3) 483-490.
  115. 115. Van Zuylen AJ, Bosman GJ, Ruitenbeek W, Van Kalmthout PJ, De Grip WJ. No evidence for reduced thrombocyte cytochrome oxidase activity in Alzheimer's disease. Neurology. 1992; 42(6) 1246-1247.
  116. 116. Adolfsson R, Gottfries CG, Oreland L, Wiberg A, Winblad B. Increased activity of brain and platelet monoamine oxidase in dementia of Alzheimer type. Life Sci. 1980; 27(12) 1029-1034.
  117. 117. Götz ME, Fischer P, Gsell W, Riederer P, Streifler M, Simanyi M, Müller F, Danielczyk W. Platelet monoamine oxidase B activity in dementia. A 4-year follow-up. Dement Geriatr Cogn Disord. 1998; 9(2) 74-77.
  118. 118. Bonuccelli U, Piccini P, Marazziti D, Cassano GB, Muratorio A. Increased platelet 3H-imipramine binding and monoamine oxidase B activity in Alzheimer's disease. J Neural Transm Park Dis Dement Sect. 1990; 2(2) 139-147.
  119. 119. Parnetti L, Reboldi GP, Santucci C, Santucci A, Gaiti A, Brunetti M, Cecchetti R, Senin U. Platelet MAO-B activity as a marker of behavioural characteristics in dementia disorders. Aging (Milano). 1994; 6(3) 201-207.
  120. 120. Zellner M, Baureder M, Rappold E, Bugert P, Kotzailias N, Babeluk R, Baumgartner R, Attems J, Gerner C, Jellinger K, Roth E, Oehler R, Umlauf E. Comparative platelet proteome analysis reveals an increase of monoamine oxidase-B protein expression in Alzheimer's disease but not in non-demented Parkinson's disease patients. J Proteomics. 2012; 75(7) 2080-2092.
  121. 121. Regland B, Gottfries CG, Oreland L. Vitamin B12-induced reduction of platelet monoamine oxidase activity in patients with dementia and pernicious anaemia. Eur Arch Psychiatry Clin Neurosci. 1991; 240(4-5) 288-291.
  122. 122. Alexopoulos GS, Young RC, Lieberman KW, Shamoian CA. Platelet MAO activity in geriatric patients with depression and dementia. Am J Psychiatry. 1987; 144(11) 1480-1483.
  123. 123. Soto J, Ulibarri I, Jauregui JV, Ballesteros J, Meana JJ. Dissociation between I2-imidazoline receptors and MAO-B activity in platelets of patients with Alzheimer's type dementia. J Psychiatr Res. 1999; 33(3) 251-257.
  124. 124. Bongioanni P, Gemignani F, Boccardi B, Borgna M, Rossi B. Platelet monoamine oxidase molecular activity in demented patients. Ital J Neurol Sci. 1997; 18(3) 151-6.
  125. 125. Danielczyk W, Streifler M, Konradi C, Riederer P, Moll G. Platelet MAO-B activity and the psychopathology of Parkinson's disease, senile dementia and multi-infarct dementia. Acta Psychiatr Scand. 1988; 78(6) 730-736.
  126. 126. Schneider LS, Severson JA, Chui HC, Pollock VE, Sloane RB, Fredrickson ER. Platelet tritiated imipramine binding and MAO activity in Alzheimer's disease patients with agitation and delusions. Psychiatry Res. 1988; 25(3) 311-322.
  127. 127. Konings CH, Scheltens P, Kuiper MA, Wolters EC. No evidence for abnormalities in kinetics of platelet monoamine oxidase in Alzheimer's disease. Clin Chim Acta. 1995; 240(1) 99-102.
  128. 128. Ahlskog JE, Uitti RJ, Tyce GM, O'Brien JF, Petersen RC, Kokmen E. Plasma catechols and monoamine oxidase metabolites in untreated Parkinson's and Alzheimer's diseases. J Neurol Sci. 1996; 136(1-2) 162-168.
  129. 129. Embi N, Rylatt DB, Cohen P. Glycogen synthase kinase-3 from rabbit skeletal muscle. Separation from cyclic-AMP-dependent protein kinase and phosphorylase kinase. Eur J Biochem. 1980; 107(2) 519-527.
  130. 130. Chin PC, Majdzadeh N, D'Mello SR. Inhibition of GSK3beta is a common event in neuroprotection by different survival factors. Brain Res Mol Brain Res. 2005; 137(1-2) 193-201.
  131. 131. Jope RS, Yuskaitis CJ, Beurel E. Glycogen synthase kinase-3 (GSK3): inflammation, diseases, and therapeutics. Neurochem Res. 2007; 32(4-5) 577-595.
  132. 132. Grimes CA, Jope RS. The multifaceted roles of glycogen synthase kinase 3beta in cellular signaling. Prog Neurobiol. 2001; 65(4) 391-426.
  133. 133. Muyllaert D, Kremer A, Jaworski T, Borghgraef P, Devijver H, Croes S, Dewachter I, Van Leuven F. Glycogen synthase kinase-3beta, or a link between amyloid and tau pathology? Genes Brain Behav. 2008; 7 Suppl 1 57-66.
  134. 134. Barry FA, Graham GJ, Fry MJ, Gibbins JM. Regulation of glycogen synthase kinase 3 in human platelets: a possible role in platelet function? FEBS Lett. 2003; 553(1-2) 173-178.
  135. 135. Forlenza OV, Torres CA, Talib LL, de Paula VJ, Joaquim HP, Diniz BS, Gattaz WF. Increased platelet GSK3B activity in patients with mild cognitive impairment and Alzheimer's disease. J Psychiatr Res. 2011; 45(2) 220-224.
  136. 136. Matsushima H, Shimohama S, Fujimoto S, Takenawa T, Kimura J. Changes in platelet phospholipase C protein level and activity in Alzheimer's disease. Neurobiol Aging. 1995; 16(6) 895-900.
  137. 137. Gattaz WF, Cairns NJ, Levy R, Förstl H, Braus DF, Maras A. Decreased phospholipase A2 activity in the brain and in platelets of patients with Alzheimer's disease. Eur Arch Psychiatry Clin Neurosci. 1996; 246(3) 129-131.
  138. 138. Gattaz WF, Forlenza OV, Talib LL, Barbosa NR, Bottino CM. Platelet phospholipase A(2) activity in Alzheimer's disease and mild cognitive impairment. J Neural Transm. 2004; 111(5) 591-601.
  139. 139. Talib LL, Yassuda MS, Diniz BS, Forlenza OV, Gattaz WF. Cognitive training increases platelet PLA2 activity in healthy elderly subjects. Prostaglandins Leukot Essent Fatty Acids. 2008; 78(4-5) 265-269.
  140. 140. Krzystanek E, Krzystanek M, Opala G, Trzeciak HI, Siuda J, Małecki A. Platelet phospholipase A2 activity in patients with Alzheimer's disease, vascular dementia and ischemic stroke. J Neural Transm. 2007; 114(8) 1033-1039.
  141. 141. Straface E, Matarrese P, Gambardella L, Vona R, Sgadari A, Silveri MC, Malorni W. Oxidative imbalance and cathepsin D changes as peripheral blood biomarkers of Alzheimer disease: a pilot study. FEBS Lett. 2005; 579(13) 2759-2766.
  142. 142. Bongioanni P, Gemignani F, Boccardi B, Borgna M, Rossi B. Platelet monoamine oxidase molecular activity in demented patients. Ital J Neurol Sci. 1997 Jun;18(3):151-156.
  143. 143. Bongioanni P, Donato M, Castagna M, Gemignani F. Platelet phenolsulphotransferase activity, monoamine oxidase activity and peripheral-type benzodiazepine binding in demented patients. J Neural Transm. 1996; 103(4) 491-501.
  144. 144. Humpel C. Identifying and validating biomarkers for Alzheimer's disease. Trends Biotechnol. 2011; 29(1) 26-32.
  145. 145. Blennow K. CSF biomarkers for Alzheimer's disease: use in early diagnosis and evaluation of drug treatment. Expert Rev Mol Diagn. 2005; 5(5) 661-672.
  146. 146. Marksteiner J, Hinterhuber H, Humpel C. Cerebrospinal fluid biomarkers for diagnosis of Alzheimer's disease: beta-amyloid(1-42), tau, phospho-tau-181 and total protein. Drugs Today (Barc). 2007; 43(6) 423-431.
  147. 147. El Haouari M, Rosado JA. Platelet function in hypertension. Blood Cells Mol Dis. 2009; 42(1) 38-43.
  148. 148. Li QX, Berndt MC, Bush AI, Rumble B, Mackenzie I, Friedhuber A, Beyreuther K, Masters CL. Membrane-associated forms of the beta A4 amyloid protein precursor of Alzheimer's disease in human platelet and brain: surface expression on the activated human platelet. Blood. 1994; 84(1) 133-142.
  149. 149. Padovani A, Pastorino L, Borroni B, Colciaghi F, Rozzini L, Monastero R, Perez J, Pettenati C, Mussi M, Parrinello G, Cottini E, Lenzi GL, Trabucchi M, Cattabeni F, Di Luca M. Amyloid precursor protein in platelets: a peripheral marker for the diagnosis of sporadic AD. Neurology. 2001; 57(12) 2243-2248.
  150. 150. Holmsen H. Signal transducing mechanisms in platelets. Proc Natl Sci Counc Repub China B. 1991; 15(3) 147-152.

Written By

Barbara Plagg and Christian Humpel

Submitted: 19 August 2014 Published: 18 November 2015