Skip to main content
Oncotarget logoLink to Oncotarget
. 2015 Oct 10;6(34):35284–35303. doi: 10.18632/oncotarget.6074

Viscoelastic and ultrastructural characteristics of whole blood and plasma in Alzheimer-type dementia, and the possible role of bacterial lipopolysaccharides (LPS)

Janette Bester 1, Prashilla Soma 1, Douglas B Kell 2, Etheresia Pretorius 1
PMCID: PMC4742105  PMID: 26462180

Abstract

Alzheimer-type dementia (AD) is a neurodegenerative disorder and the most common form of dementia. Patients typically present with neuro- and systemic inflammation and iron dysregulation, associated with oxidative damage that reflects in hypercoagulability. Hypercoagulability is closely associated with increased fibrin(ogen) and in AD patients fibrin(ogen) has been implicated in the development of neuroinflammation and memory deficits. There is still no clear reason precisely why (a) this hypercoagulable state, (b) iron dysregulation and (c) increased fibrin(ogen) could together lead to the loss of neuronal structure and cognitive function. Here we suggest an alternative hypothesis based on previous ultrastructural evidence of the presence of a (dormant) blood microbiome in AD. Furthermore, we argue that bacterial cell wall components, such as the endotoxin lipopolysaccharide (LPS) of Gram-negative strains, might be the cause of the continuing and low-grade inflammation, characteristic of AD. Here, we follow an integrated approach, by studying the viscoelastic and ultrastructural properties of AD plasma and whole blood by using scanning electron microscopy, Thromboelastography (TEG®) and the Global Thrombosis Test (GTT®). Ultrastructural analysis confirmed the presence and close proximity of microbes to erythrocytes. TEG® analysis showed a hypercoagulable state in AD. TEG® results where LPS was added to naive blood showed the same trends as were found with the AD patients, while the GTT® results (where only platelet activity is measured), were not affected by the added LPS, suggesting that LPS does not directly impact platelet function. Our findings reinforce the importance of further investigating the role of LPS in AD.

Keywords: Alzheimer-type dementia, iron levels, lipopolysaccharides, scanning electron microscopy, thromboelastography®, Gerotarget

INTRODUCTION

Alzheimer-type dementia (AD) is a neurodegenerative disorder and the most common form of dementia [13]. Dementia is a syndrome applied to a group of symptoms that can be caused by a variety of conditions, the most common of which is Alzheimer's disease; unfortunately, due in part to this ambiguity, the aetiology of AD is not well understood [4]. The onset and risk of -development is still mostly unexplained, and only very partially so by genetic factors [4]. Today AD is the largest unmet medical need in neurology [57], and is a condition characterized by neuroinflammation [8, 9]. Neuroinflammation is a multi-faceted and complex phenomenon where the precise mechanisms have not been completely elucidated [10], but where activated astrocytes and microglia are usually the trigger in the neuroinflammatory process. They become reactive in response to virtually all pathological situations in the brain such as axotomy (neuritic dystrophy), ischemia, infection, and existing neurodegenerative diseases [1115].

In this paper, we discuss the factors involved in systemic inflammation and show that they are also typically prevalent in neuroinflammation. We focus specifically on literature that shows a changed iron profile, increased fibrinogen levels and oxidative stress; these have each been implicated in a typical hypercoagulable (thrombotic) state, but are also present in AD (the literature is discussed fully in the different sections). Figure 1 shows the layout of this paper, where the focus is on 3 main characteristics of AD, namely (1) neuroinflammation (2) systemic inflammation and (3) oxidative stress. We briefly discuss specific changes in brain morphology (A) and shared inflammatory mediators between neuroinflammation and systemic inflammation (B) (with special reference to a changed iron profile). Factors that are typically implicated in systemic inflammation, but that have an impact on the brain (C) are then examined. We then consider how the three main characteristics are affected by and also result in (4) hypercoagulation and how we can measure this by using electron microscopic and viscoelastic techniques. Lastly, we conclude by discussing possible reasons for this hypercoagulability in AD, and we discuss that the presence of a dormant blood microbiome, and in particular the lipopolysaccharides (LPS) that they can shed, may be central to the hypercoagulable state in this condition [16, 17].

Figure 1. Alzheimer-type dementia (AD) and three of its main characteristics (1) neuroinflammation (2) systemic inflammation and (3) oxidative stress, where A.

Figure 1

involves specific changes in brain morphology and B. shows shared inflammatory mediators between neuroinflammation and systemic inflammation (with special reference to a changed iron profile) and C. factors that are typically implicated in systemic inflammation, but that have an impact on the brain. The three main characteristics are affected by and also result in (4) hypercoagulation, which we can measure using electron microscopy and viscoelastic techniques. We conclude by discussing how the presence of a dormant blood microbiome, and in particular the lipopolysaccharides (LPS) and other cell wall materials that they can shed, may be central to the hypercoagulable/neuroinflammatory state in this condition.

NEUROINFLAMMATION AND THE IRON LINK

The next paragraphs will briefly discuss presentation of neuroinflammation and the involvement of iron and its accompanying oxidative damage in AD etiology. We will make the link between the presence of hypercoagulability in AD and oxidative damage, changed iron levels and inflammation (both neuro- and systemic inflammation).

The hallmarks of dementia in AD, as well as the severity and state of neurodegeneration, are closely linked to the presence of tau phosphorylation, amyloid-β peptide aggregation, neurofibrillary tangle formation, neuroinflammation, and neurodegeneration [18, 19]. If we look closely at the presentation of AD, it is characterized by brain lesions known as intracellular neurofibrillary tangles and extracellular neuritic plaques surrounded by activated astrocytes and microglia [20]. The neurofibrillary tangles consist of paired helical filaments of truncated tau protein that is abnormally hyperphosphorylated [21, 22]. The main component in the plaques is the amyloid-β peptide [1, 23], though its role in the actual disease pathology is more than questionable [24]. The brain lesions in AD are also characterized by the presence of a broad spectrum of inflammatory mediators (complement proteins, inflammatory cytokines, prostaglandins, and acute phase reactants such as C-reactive protein and amyloid P) [25, 26]. Resident brain cells, including neurons produce these mediators [27, 28]. Neuroinflammation is therefore key in AD [26, 2833]. Central to this neuroinflammation in AD is the involvement of iron and its accompanying oxidative damage in AD etiology [3448]. Oxidative damage is one of the earliest pathological changes in AD, and the aberrant redox activity is therefore also among the earliest changes in the transition to the disease state [49]. Note that as long ago as 1991 it was shown that chelators of free iron improved cognitive function in AD sufferers [40, 50].

OXIDATIVE DAMAGE, IRON, INFLAMMATION AND HYPERCOAGULABILITY

Oxidative damage, increased iron levels and inflammation are all linked to the development of hypercoagulability [5158]. Increased fibrin(ogen) levels have also been noted in hypercoagulation [53, 5965] and this is also observed in blood vessels positive for amyloid in mouse and human AD samples [66]. Fibrinogen extravasation in the AD brain has also been noted [67]. Thus, fibrin(ogen) may play an important role in AD etiology as it has been implicated in the neuroinflammation, neurovascular damage, blood-brain barrier permeability, vascular amyloid deposition, and memory deficits, all of which are associated with AD [68]. Increased fibrinogen levels in AD are therefore a strong cerebrovascular risk factor in these patients, as fibrinogen specifically binds to β-amyloid, thereby altering fibrin clot structure and delaying clot degradation [69]. However, there is currently no evidence regarding the extent to which delayed clot degeneration might contribute to AD.

Thrombosis, and therefore hypercoagulation, is typically associated with increased levels of fibrin(ogen) [58, 70, 71]. A changed iron profile (e.g. low transferrin and/or high serum ferritin) is also central to thrombosis and hypercoagulation [38, 58, 7279] and iron dysregulation is heavily implicated in AD [38, 80]. Changes in fibrin fibre structure (due to hypercoagulation and a changed iron profile) can be visualized using various ultramicroscopy techniques [54, 58, 8186]. In addition, these changes might be correlated with results from viscoelastic techniques like thromboelastography (TEG®) [52, 79, 85, 87, 88]. Figure 2 shows how the intrinsic and extrinsic coagulation process causes fibrin fibre formation under normal conditions, and how fibrin fibre formation is changed under the influence of a changed (aberrant) iron profile, whether by generating hydroxyl radicals (resulting in oxidative stress, and also during systemic inflammation) [89] or by electrostatic means [57, 58].

Figure 2. Schematic representation of the intrinsic and extrinsic coagulation pathway and the conversion of soluble fibrinogen into insoluble polymers.

Figure 2

Thrombin‘s action on fibrinogen results in the formation of fibrin strands (left insert) that are in time degradable with plasmin. By contrast, iron-induced dense matted deposits (right insert) are less degradable [89] - this is during normal blood coagulation. Iron-induced blood coagulation is seen as dense matted deposits under scanning electron microscopy and viscoelastically [85, 87, 88].

LINK BETWEEN A HYPERCOAGULABLE STATE, IRON DYSREGULATION, CHANGES IN FIBRIN FIBRE STRUCTURE AND A STERILE MICROBIOME

This said, there is still no clear reason precisely why (a) this hypercoagulable state, (b) iron dysregulation and (c) changes in fibrin fibre structure could together lead to the loss of neuronal structure and cognitive function in AD. The following paragraphs will discuss the link between (a to c) and a supposedly sterile blood microbiome.

Recently, we showed (with ultrastructural microscopy techniques) that, in the “sterile” blood microbiome of AD patients, there are bacteria present, that we have suggested are normally dormant [16, 90]. Dormancy is in fact the norm in environmental and general microbiology [91, 92]. We note that there is a substantial literature implicating chronic infection [9397] as well as the gastrointestinal tract microbiome in AD [98100]. In particular, periodontal disease related pathogens and their inflammatory products have been shown to contribute to, or at least to accompany, systemic inflammation and the pathogenesis of AD [93, 101105]. Particularly, periodontal pathogens Porphyromonas gingivalis, Tannerella forsythia, and Treponema denticola have been implicated in the development of AD [106, 107]. Infections with Herpes simplex virus type 1 [97], picornavirus, Borna disease virus, Chlamydia pneumoniae, Helicobacter pylori, and spirochetes [96, 108] and pathogens causing urinary tract infections [94, 109111] are also implicated and co-occurring in AD.

Recently, immunoblotting demonstrated bands corresponding to lipopolysaccharides (LPS) (also known as endotoxin), produced or shed by P. gingivalis in 40% of AD brain specimens [107]. Indeed, there is evidence that bacterial endotoxins are directly involved in the inflammatory and pathological processes associated with AD [112]. Interestingly, it has been observed that chronic infusion of the bacterial LPS, the outer cell wall component of Gram negative bacteria, into the fourth brain ventricle of rats reproduces many of the inflammatory and pathological features seen in the brain of AD patients [112]. Apolipoprotein E (ApoE) is an intermediate-density lipoprotein that is essential for the normal catabolism of triglyceride-rich lipoprotein constituents. It also transports cholesterol (its primary function), and regulates amyloid-β (Aβ) metabolism, aggregation, and deposition [113]. Interestingly, LPS may disturb the typically anti-inflammatory effect of ApoE. ApoE function is known to be down-regulated in AD patients [114, 115], and carrying the ApoE4 allele increases risk of Alzheimer's disease [113, 116, 117]. The down-regulating effect on ApoE by LPS was also seen in an animal model [118]. Furthermore, LPS is used in animal models to induce AD-like symptoms, as well as neuroinflammation [119, 120], as well as Parkinson's disease-like symptoms [17, 121128]. Recently it was also noted that LPS can produce myelin injury and plaque-like aggregates of myelin in mice, and amyloid-β and amyloid-β protein precursor co-localize with these myelin aggregates. Cortical amyloid plaques also co-localized with myelin aggregates [129]. Following LPS injection, Alzheimer-like amyloidogenic axonal pathology also occur in the normal mammalian brain in partnership with neuroinflammation [130], and LPS is associated with the development of neuroinflammation in animals [130], as well as in primary culture neuroinflammatory models [131].

From the above, and other literature reviewed elsewhere [17, 132], there is ample evidence that LPS can cause neuroinflammation and amyloid-β formation. There is also evidence for the involvement of LPS in cognitive impairment in AD. For instance,

  • LPS is used to induce cognitive impairment in mice [120, 133].

  • LPS induces stress and depression in late onset AD [134].

  • Neuro-inflammation, amyloidogenesis and memory impairment in a mouse model is seen following the systemic inflammation generated by LPS [120].

  • Intraperitoneal injection of LPS causes attention deficits [135] and severe and fluctuating cognitive deficits in 16-week ME7 mice [136].

Where does this bacteria/LPS argument lead us to? It is known that LPS can also cause hypercoagulation [137, 138]; this has been referred to as endotoxin-mediated hypercoagulation [139]. There is substantial evidence in the literature that microbes have a prominent involvement in AD, but it seems that only a few researchers have taken the final step to suggest that such bacteria may actually cause or exacerbate AD (in the sense of “Koch's postulates”) [96], and that shedding of LPS from the (dormant) blood microbiome may exacerbate or even cause the hypercoagulability seen in AD. Clearly the measurement of hypercoagulability is considerably easier than the estimation of cognitive function.

In the current paper we used scanning electron microscopy, together with viscoelastic techniques, to study whole blood and plasma in AD patients and compared the results with those of age- and gender-matched healthy individuals. We also studied iron profiles and general hematological parameters. Lastly, we simulated the effect of “physiological” levels of LPS, by adding it to healthy, uncitrated blood. We determine if LPS has an effect on platelets by using a novel technique called the Global Thrombosis Test (GTT) [140, 141]. This allows for the detection of thrombin generation by activated platelets, the major determinant of arterial thrombogenesis and measured endogenous (spontaneous) thrombolytic activity (http://www.globalthrombosis.com). Typical LPS concentrations in ‘normal’ whole blood in healthy subjects seem to be of the order of 10-15 ng.L−1 [17, 142, 143], while those of LBP are roughly 1,000,000 times greater at 5-15 mg.L−1 (with both values increasing during sepsis). The exact significance of these numbers is not clear [17] as LPS is so hydrophobic that most is bound to the LPS-binding protein or lipoproteins [144146]. We also added LPS to naïve whole blood and measured coagulation parameters with the TEG®, to determine directly if added LPS causes hypercoagulability. If this were to be the case, it would imply that LPS can bind directly to fibrinogen.

RESULTS

Healthy individual and Alzheimer-type dementia patient data

Table 2 show demographics for healthy and AD individuals, iron profiles, and data for the TEG®, GTT® and SEM.

Table 2. Demographics of participants, as well as iron levels, thromboelastography® (TEG®) of plasma, fibrin fiber thickness, and thromboelastography® (TEG®) and global thrombosis test (GTT®) of naïve blood of age-matched controls (without dementia) and Alzheimer-type dementia (AD) patients, showing medians, standard deviation and p-values (values lower than 0.05 are indicated in blue) obtained using the Mann-Whitney U test.

Variables Healthy individuals (n = 20) Alzheimer-type dementia individuals (n = 40) P-value Confidence interval (95%) relative to median
AGE years 68.5 (±22.21) 79 (±11.8) 0.316 −4 to 21
GENDER
Male 6 (40%) 12 (30%)
Female 14 (60%) 28 (70%)
IRON PROFILES
Iron μM 18.8 (± 5.74) 13.55 (±4.98) 0.0293 0.4 to 6.8
Transferrin g.L−1 2.55 (± 0.39) 2.11 (± 0.39) 0.0001 0.2 to 0.7
% Saturation 28 (± 10.32) 26 (± 9.95) 0.6543 −5 to 8
Serum Ferritin ng.mL−1 79 (±78.39) 99 (±128.84) 0.457 −59 to 21
THROMBOELASTOGRAPHY® OF PLATELET POOR PLASMA
MRTG 5.35 (± 2.83) 8.47 (± 4.82) 0.0013 1.21 to 4.79
TMRTG 9.46 (± 4.24) 7.79 (± 5.02) 0.0428 0.08 to 2.67
TTG 243.89 (± 85.66) 283.97 (± 116.51) 0.157 −97.47 to 16.3
R 7.2 (± 3.60) 6.2 (± 4.18) 0.122 −0.2 to 2
K 2.35 (± 0.73) 1.4 (± 4.23) 0.0015 0.3 to 1.2
Angle 66.85 (± 9.60) 74.05 (± 8.64) < 0.0001 −10.6 to −3.9
MA 32.7 (± 8.96) 36.1 (± 9.67) 0.15 −8.3 to 1.4
FIBRIN FIBRE THICKNESS n = 1000 n = 2000
Fibre thickness in nm 110 (± 33.8) 115 (± 44.18) <0.0001 6 to 12
TEG® RESULTS OF NAÏVE WHOLE BLOOD WITH AND WITHOUT ADDED LPS
Healthy Whole Blood(n = 10) Healthy Whole Blood with added LPS(n = 10)
MRTG 2.42 (± 0.41) 2.835 (± 0.67) 0.404 −0.82 to 0.44
TMRTG 15.04 (± 2.98) 11.835 (± 1.92) 0.0003 1.66 to 6.25
TTG 605.21 (± 122.53) 620.89 (± 133.94) 0.739 −135.27 to 81.88
R 10.2 (± 1.01) 7.1 (± 1.82) 0.001 1.8 to 3.9
K 5.35 (± 1.08) 5 (± 1.28) 0.252 −0.6 to 1.7
ANGLE 44.8 (± 4.86) 49.1 (± 2.08) 0.085 −6.5 to 1.1
MA 53.65 (± 5.80) 55 (± 5.13) 0.616 −4.9 to 3.1
GTT® RESULTS OF NAÏVE WHOLE BLOOD WITH AND WITHOUT ADDED LPS
Healthy Whole Blood(n = 10) Healthy Whole Blood with added LPS(n = 10)
OT 335.8 (± 80.96) 354.35 (± 73.66) 0.481 −101.8 to 49
LT 1576 (± 322.24) 1534.5 (± 155.19) 0.315 −93 to 369

Literature suggests that normal values for serum iron are between 11.6 and 31.4 μmol.L−1 [158], for transferrin they are 2.2 - 3.7 g.L−1 [158], normal % saturation is 20-50% [158], and serum ferritin for males are between 25 - 300 μg.L−1 and females are between 25 - 200 μg.L−1 [159]. However, values for normal ranges vary between pathology laboratories. Our values are therefore taken according to the values for South Africa (http://ampath.co.za) and indicated in the table. In the current sample, the iron and transferrin showed significant differences between the healthy and AD population, with the transferrin for the AD individuals being mostly lower (see Table 2).

In our previous paper on the hematological system and systemic inflammation of AD, we reviewed the importance of increased iron, and particularly serum ferritin [160]. Results from the current AD sample show that both iron and transferrin levels were significantly different between the healthy and AD individuals. Transferrin is lowered in inflammation [72, 161], and is known as a “negative” acute-phase protein [161]; it is also decreased in in AD [162]. In the present sample most of the AD individuals have serum ferritin levels within the normal ranges for their genders.

Thromboelastography® results

The following parameters were all changed in the AD patients, and showed a significant difference between the healthy and AD individuals:

  • Increased maximum rate of thrombus generation (MRTG),

  • Decreased time to maximum rate of thrombus generation (TMRTG) which is the time interval (s) observed before maximum velocity of clot growth,

  • Shorter time taken to achieve final clot strength i.e. amplification (K)

  • as well as an increased Alpha or Angle representing the speed at which the fibrin build-up and crosslinking of the fibrinogen fibres takes place, or therefore to stabilize the clot.

The results are in line with a faster initial clot formation but and increased time to stabilize the clot in AD compared to controls.

Scanning electron microscopy (SEM) of whole blood and platelet poor plasma (PPP) results

Due to the observations that the viscoelastic properties of clot generation were significantly changed as judged using the TEG®, SEM was performed with both whole blood and PPP. Figure 3 shows SEM micrographs from whole blood and PPP of healthy individuals, while Figures 4 and 5 shows examples from AD individuals. Whole blood from healthy individuals typically shows discoid erythrocytes (RBCs), with platelets that are typically discoid, without pseudopodia and a few that are slightly activated, usually due to contact activation [53, 160, 163165] (Figure 3A). This slight activation, with resulting pseudopodia formation, is due to contact activation [166]. This ultrastructure is in line with our database of thousands of micrographs from healthy individuals. Although, for the current study we used age- and gender-matched healthy individuals, there are not great variations between RBCs and fibrin networks of young and old individuals when they are healthy. RBCs are typically discoid and fibrin fibres typically are seen as individual strands.

Figure 3.

Figure 3

Whole blood smear showing a typical erythrocyte; Scale bar: 1 μm A.; two healthy platelets with slight pseudopodia formation; Scale bar: 300 nm B.; and an extensive fibrin network created by adding thrombin (58 nM thrombin final concentration with plasma) to platelet poor plasma; Scale bar: 1 μm C.

Figure 4. Whole blood smear showing erythrocyte and platelet interactions from Alzheimer-type dementia individuals, with bacterial presence.

Figure 4

A. to F. Hyperactivated platelets with spreading are shown in G. and H. White arrows show platelet spreading, indicative of inflammation and hyperactivation - this is known to happen in all inflammatory conditions; for a discussion on hyperactivation of platelets see [166]. Blue arrows show matted plasma/fibrin deposits. False yellow colouring was added to emphasize the presence of microbiota. Red block shows eryptotic erythrocyte. P shows partially intact but activated platelets, where spreading is starting happen, and white block surrounds such an activated platelet mass. Scale bars: 1 μm.

Figure 5. Extensive fibrin networks from two healthy individuals (A and B) and from two Alzheimer-type dementia individuals (C and D) created by adding thrombin to platelet poor plasma.

Figure 5

White arrow possibly shows LPS or coagulated plasma proteins, red arrow and red line shows thicker fibrin fiber. Scale bar: 1 μm

Figures 4 and 5 show extensive fibrin networks from whole blood and PPP for AD individuals. Recently we noted the presence of bacteria inside and around RBCs of AD patients [90]. Figure 4D also shows an example of an eryptotic erythrocyte. The term eryptosis is a type of suicidal death of erythrocytes, that was discovered recently. It is characterized by erythrocyte shrinkage, blebbing, and phospholipid scrambling of the cell membrane. For a detailed discussion on eryptosis see various publications by Lang and Quadri [167] [168] [169, 170]. Previously it was reported in AD, that Amyloid peptides may induce RBC eryptosis.

In the current, newly collected samples we also found this phenomenon in whole blood of all individuals from our AD sample (Figure 4). Due to the descriptive nature of SEM analysis, any kind of exact estimation of bacterial numbers per volume blood is unfortunately not possible with this method. Platelets were overactivated and showed extreme spreading, as typically seen during systemic inflammation [166, 171, 172].

Due to the presence of (visible) bacteria in whole blood smears, (as seen with electron microscopy) from AD individuals and therefore potentially the presence of cell wall material that they shed, LPS may be one of the culprits, or at least contribute to the hypercoagulability in AD.

Figure 5 shows fibrin fibre networks from two healthy individuals (A and B) and two AD individuals; the networks are created by adding thrombin to plasma. We also performed measurements on the fibrin fibre diameters using ImageJ (ImageJ is a public domain, Java-based image processing program developed at the National Institutes of Health: http://rsbweb.nih.gov/ij/); Table 2 shows the median fibrin fibre diameter and statistical analysis for the healthy and AD individual fibrin fibres (see Figure 6). Previously we noted a significant difference in fibrin fibre thickness in a pilot study where we measured fibrin fibre thickness in platelet rich plasma (PRP) from AD patients with normal and high serum ferritin values [88]. With regard to median fibrin fibre width, the value for the healthy (younger) subjects was 105± 3 nm [58]. These measurements were taken on samples from younger individuals. The high ferritin AD group had a fibre width that was significantly higher than the normal ferritin AD group, with 34% less than the high ferritin group [88]. The current results showed that the median fiber thickness of older healthy individuals was 110 nm (± 34 nm) and the median AD fibre thickness was 115 nm (± 44 nm). There seems to be a linkage between fibre diameter and serum ferritin levels, which were not different in the present samples.

Figure 6. Fibrin fibre thickness (nm) distribution width for both healthy A.

Figure 6

and Alzheimer-type dementia individuals B.

GTT® and TEG® analysis with added LPS

Due to the presence of bacteria in the whole blood of AD individuals as seen in the SEM images, we simulated the potential presence of LPS in naïve whole blood from healthy individuals by adding it. A very low final concentration of 0.2 ng.L−1 caused a hypercoagulable state using the TEG® in naïve whole blood when the LPS was added. The TEG® measures the activity of the whole coagulation pathway. The following two parameters were significantly changed to a more hypercoagulable state in when LPS was added to the blood of the healthy individuals:

R value (reaction time in seconds) is shorter with added LPS, showing the time of latency from start of test to initial fibrin formation is decreased.

Time to maximum rate of thrombus generation (TMRTG) is also shorter with added LPS, showing a lowering of the time interval in seconds before maximum velocity of clot growth.

This suggests that added LPS causes quicker and firmer clot generation.

Results from the GTT® showed that added LPS did not have an effect on platelet shear force in the GTT® (seen Table 2). As mentioned previously, this GTT® test measures platelet reactivity (occlusion time, OT), where an OT of less than 300 seconds indicated platelet hyper-reactivity, while one between 300 and 500 indicates normal haemostatic/platelet activity [140]. The median of the OT of the healthy individuals with and without added LPS were in the normal ranges, suggesting that LPS do not affect platelet activity. The GTT also shows the time to lyse (LT), and normal values are less than 2000 seconds. Table 2 shows the median for the healthy individuals are less than 2000 seconds, and that LPS does not cause low thrombotic activity.

One interpretation is that LPS possibly binds (due to its very lipophilic nature) to plasma proteins involved in the coagulation cascade, resulting in the hypercoagulability of naïve whole blood with added LPS. This is supported by the TEG® results. However, measuring OT and LT with the GTT, we see no significant changes between naïve whole blood with and without added LPS (see Table 2). This suggests that LPS does not affect platelet activity.

DISCUSSION

In the current sample we see that the following parameters were significantly different between healthy and Alzheimer-type dementia individuals:

  • Iron (free, is less in AD)

  • Transferrin (is less in AD)

  • Maximum rate of thrombus generation (MRTG),

  • Time to maximum rate of thrombus generation (TMRTG) which is the time interval (s) observed before maximum velocity of clot growth,

  • Time taken to achieve final clot strength i.e. amplification (K)

  • Alpha or Angle representing the speed at which the fibrin build-up and crosslinking of the fibrinogen fibres takes place.

  • Fibrin fibre thickness

These results are indicative of increased coagulability in AD, suggesting structural changes in the physical fibrin fibre packaging. The presence of bacteria in whole blood in AD individuals, as seen with SEM as shown in this paper, suggest that LPS, known to be shed from (Gram-negative) bacterial membranes may play a role in the increased coagulability that is seen in AD PPP.

We assessed coagulation of naive blood with and without LPS. TEG® of naïve healthy blood with added LPS showed that it was indeed the case that LPS affects coagulability, as both the

  • R value and

  • Time to maximum rate of thrombus generation (TMRTG)

showed increased coagulability (due to a shorter time to maximum rate of thrombus generation) and also shorter initial clot formation, where R is also shorter with added LPS. This suggests that added LPS causes quicker and firmer clot generation. This is in line with the hypercoagulability seen in AD individuals. Due to the very variable and often unreliable currently available measurements for LPS in whole blood (for a review see [17], basal LPS levels were not measured in the samples. GTT, which measures only platelet activity, was not affected by the added LPS. These results suggest that LPS does not directly impact platelet function (at least in a controlled experimental environment).

LPS is known to induce inflammation via cytokine activation [173175], and a characteristic of inflammation is almost always a hypercoagulatory state [58, 176183]. LPS may also cause hypercoagulation via tissue factor-(TF-) mediated activation of hemostasis in whole blood samples from adults and neonates [184]. TF is also a cytokine. We suggest that there is a third possible route of acute activation by directly binding to plasma proteins involved in the coagulation cascade to cause hypercoagulation. The results presented here support such an acute (fast) reaction.

These results suggest that the clot in AD forms faster, resulting in a firmer clot, reflected in both a decreased time to clot formation and time to maximum firmness of clot. The reason for this changed fibrin structure may be due to a changed fibrinogen packaging during the clot formation, and this changed packaging may be due to the presence of LPS. Future work will seek to establish this directly.

MATERIALS AND METHODS

Volunteer details and blood collection

Blood samples were obtained from non-smoking Alzheimer-type dementia (AD) patients, identified by a Neurologist and under the care of a medical practitioner. Specifically, care was taken to exclude vascular dementia. We also recruited “healthy” age-matched individuals that did not smoke. It should be noted that the term “healthy” is used in this paper to describe an individual that has does not have dementia. Ethical clearance was obtained from the Health Sciences Ethical committee from the University of Pretoria, and informed consent was obtained from family members who act as carers of the patients. Healthy individuals also filled in consent forms. Blood was collected in two x 4mL citrate tubes, one EDTA tube, and one 4mL clotting tube for iron level determination. This collection and all handling of samples were performed under very strictly aseptic conditions, in order to prevent any microbial contamination of samples.

Statistical analysis

The non-parametric Mann-Whitney U test was adopted to determine P-values using the software StatsDirect (www.statsdirect.com). A p-value of less than 0.05 was considered statistically significant (but cf. e.g. [147, 148]).

Iron tests

Serum ferritin was measured by using the Bio-Rad Laboratories’ QuantImune ferritin IRMA kit that is a single-incubation two-site immunoradiometric assay. In this IRMA, which measures the most basic isoferritin, the highly purified I-labeled antibody to ferritin is the tracer and the ferritin antibodies are immobilized on polyacrylamide beads as the solid phase. Transferrin was measured with the RayBio® Human Transferrin ELISA from RayBiotech. Serum iron was measured with the Iron Assay kit (Colorimetric assay).

Viscoelastic tests using platelet poor plasma (PPP)

Coagulation parameters, using PPP of patients and healthy individuals, were done using thromboelastography® (TEG®). PPP was brought to room temperature and 340 μl was placed in a disposable cup in a computer-controlled TEG® hemostasis system (Model 5000, Hemoscope, Niles, IL), with addition of 20 μl CaCl2 as the last step to initiate clotting. Thrombelastographic data were collected until maximum elastic modulus (MG) is reached or 60 min had elapsed [79, 88, 149152]. See Table 1 for the parameters that can be obtained when both plasma and whole blood are studied using the TEG®. TEG® is typically used to determine clot formation and clot strength [85].

Table 1. Thromboelastograph® parameters typically generated for whole blood and platelet poor plasma [153, 154].

THROMBOELASTIC PARAMETERS
R value: reaction time Minutes Time of latency from start of test to initial fibrin formation (amplitude of 2mm); i.e. initiation time
K: kinetics Minutes Time taken to achieve a certain level of clot strength (amplitude of 20mm); i.e. amplification
A (Alpha): Angle (slope between the traces represented by R and K) Angle in degrees The angle measures the speed at which fibrin build up and cross linking takes place, hence assesses the rate of clot formation; i.e. thrombin burst
MA: Maximal Amplitude mm Maximum strength/stiffness of clot. Represents the ultimate strength of the fibrin clot, i.e. overall stability of the clot
Maximum rate of thrombus generation (MRTG) Dyn.cm−2.s−1 The maximum velocity of clot growth observed
Time to maximum rate of thrombus generation (TMRTG) Minutes The time interval observed before the maximum speed of the clot growth
Total thrombus generation (TTG) Dyn.cm−2 The clot strength: the amount of total resistance (to movement of the cup and pin) generated during clot formation. This is the total area under the velocity curve during clot growth representing the amount of clot strength generated during clot growth

Scanning electron microscopy (SEM) of whole blood and platelet poor plasma (PPP)

At least 30 minutes after the blood was collected, 10 μl of whole blood were placed directly on a glass cover slip, fixed, dehydrated, dried, mounted and coated with carbon according to previously described methods [155]. Platelet poor plasma (PPP) were obtained and frozen at −80°C. After all samples were collected, PPP were thawed and 10 μl mixed with 5 μl thrombin to create an extensive fibrin network. A Zeiss ULTRA Plus FEG-SEM with InLens capabilities was used to study the surface morphology of erythrocytes, and micrographs were taken at 1kV.

The Global thrombotic test (GTT®) and thromboelastography® (TEG®) on naïve, uncitrated whole blood with and without added LPS (final LPS concentration 0.2 ng.L-1)

As mentioned above, this GTT® test measures platelet reactivity (occlusion time, OT), where an OT of less than 300 seconds indicated platelet hyper-reactivity, while one of between 300 and 500 indicates normal haemostatic/platelet activity [140]. It also shows the time to lyse or lysis time (LT), where an LT of less than 2000 seconds shows a normal spontaneous thrombolytic activity and LT of 2000 to 4000 seconds shows a reduced thrombolytic activity [156]. Due to the physical constraints of this test (naïve, uncitrated blood is drawn and immediately dispensed into the machine, and also mixed with LPS (final concentration: 0.2 ng.L−1 LPS), and incubated for 3 minutes, only the blood of 10 healthy individuals who came into the laboratory for the tests, were used. LPS from Escherichia coli O111:B4 was purchased from Sigma, product number L 2630. Naïve and LPS-treated naïve healthy blood was also tested with the TEG®, without added CaCl2. These tests were done to determine if LPS causes either (1) platelet-induced hypercoagulability or (2) fibrinogen-induced hypercoagulability in healthy blood. This was done to simulate LPS effects, as our hypothesis states that the hypercoagulability seen in AD, together with the presence of increased serum ferritin and low transferrin levels [157], is related to the presence of bacteria in sterile blood, and that these bacteria are visible using SEM.

Acknowledgments

We thank the Biotechnology and Biological Sciences Research Council (grant BB/L025752/1) as well as the National Research Foundation (NRF) of South Africa for supporting this collaboration. This is also a contribution from the Manchester Centre for Synthetic Biology of Fine and Speciality Chemicals (SYNBIOCHEM) (BBSRC grant BB/M017702/1).

Footnotes

CONFLICTS OF INTEREST

The authors have no conflict of interests to declare.

REFERENCES

  • 1.Meraz-Rios MA, Toral-Rios D, Franco-Bocanegra D, Villeda-Hernandez J, Campos-Pena V. Inflammatory process in Alzheimer's Disease. Frontiers in integrative neuroscience. 2013;7:59. doi: 10.3389/fnint.2013.00059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Zou Z, Liu C, Che C, Huang H. Clinical genetics of Alzheimer's disease. BioMed research international. 2014;2014:291862. doi: 10.1155/2014/291862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kim DH, Yeo SH, Park JM, Choi JY, Lee TH, Park SY, Ock MS, Eo J, Kim HS, Cha HJ. Genetic markers for diagnosis and pathogenesis of Alzheimer's disease. Gene. 2014;545(2):185–193. doi: 10.1016/j.gene.2014.05.031. [DOI] [PubMed] [Google Scholar]
  • 4.Morris JK, Honea RA, Vidoni ED, Swerdlow RH, Burns JM. Is Alzheimer's disease a systemic disease? Biochimica et biophysica acta. 2014;1842(9):1340–1349. doi: 10.1016/j.bbadis.2014.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Skaper SD. Alzheimer's disease and amyloid: culprit or coincidence? International review of neurobiology. 2012;102:277–316. doi: 10.1016/B978-0-12-386986-9.00011-9. [DOI] [PubMed] [Google Scholar]
  • 6.Fargo KN, Aisen P, Albert M, Au R, Corrada MM, DeKosky S, Drachman D, Fillit H, Gitlin L, Haas M, Herrup K, Kawas C, Khachaturian AS, Khachaturian ZS, Klunk W, Knopman D, et al. 2014 Report on the Milestones for the US National Plan to Address Alzheimer's Disease. Alzheimer's & dementia : the journal of the Alzheimer's Association. 2014;10(5 Suppl):S430–452. doi: 10.1016/j.jalz.2014.08.103. [DOI] [PubMed] [Google Scholar]
  • 7.Association As Alzheimer's disease facts and figures. Alzheimer's & dementia : the journal of the Alzheimer's Association. 2015;11(3):332–384. doi: 10.1016/j.jalz.2015.02.003. 2015. [DOI] [PubMed] [Google Scholar]
  • 8.Thériault P, ElAli A, Rivest S. The dynamics of monocytes and microglia in Alzheimer's disease. Alzheimer's research & therapy. 2015;7(1):41. doi: 10.1186/s13195-015-0125-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.ElAli A, Rivest S. Microglia in Alzheimer's disease: A multifaceted relationship. Brain, behavior, and immunity. 2015 doi: 10.1016/j.bbi.2015.07.021. [DOI] [PubMed] [Google Scholar]
  • 10.Steardo L, Jr, Bronzuoli MR, Iacomino A, Esposito G, Steardo L, Scuderi C. Does neuroinflammation turn on the flame in Alzheimer's disease? Focus on astrocytes. Frontiers in neuroscience. 2015;9:259. doi: 10.3389/fnins.2015.00259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ben Haim L, Carrillo-de Sauvage MA, Ceyzériat K, Escartin C. Elusive roles for reactive astrocytes in neurodegenerative diseases. Frontiers in cellular neuroscience. 2015;9:278. doi: 10.3389/fncel.2015.00278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.von Bernhardi R, Eugenín-von Bernhardi L, Eugenín J. Microglial cell dysregulation in brain aging and neurodegeneration. Frontiers in aging neuroscience. 2015;7:124. doi: 10.3389/fnagi.2015.00124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wang WY, Tan MS, Yu JT, Tan L. Role of pro-inflammatory cytokines released from microglia in Alzheimer's disease. Annals of translational medicine. 2015;3(10):136. doi: 10.3978/j.issn.2305-5839.2015.03.49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Denes A, Thornton P, Rothwell NJ, Allan SM. Inflammation and brain injury: acute cerebral ischaemia, peripheral and central inflammation. Brain, behavior, and immunity. 2010;24(5):708–723. doi: 10.1016/j.bbi.2009.09.010. [DOI] [PubMed] [Google Scholar]
  • 15.Drake C, Boutin H, Jones MS, Denes A, McColl BW, Selvarajah JR, Hulme S, Georgiou RF, Hinz R, Gerhard A, Vail A, Prenant C, Julyan P, Maroy R, Brown G, Smigova A, et al. Brain inflammation is induced by co-morbidities and risk factors for stroke. Brain, behavior, and immunity. 2011;25(6):1113–1122. doi: 10.1016/j.bbi.2011.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kell DB, Potgieter M, Pretorius E. Individuality, phenotypic differentiation, dormancy and ‘persistence’ in culturable bacterial systems: commonalities in environmental, laboratory, and clinical microbiology. F1000Review. 2015 doi: 10.12688/f1000research.6709.1. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kell DB, Pretorius E. On the translocation of bacteria and their lipopolysaccharides between blood and peripheral locations in chronic, inflammatory diseases: the central roles of LPS and LPS-induced cell death. Integrative Biology. 2015 doi: 10.1039/c5ib00158g. online. [DOI] [PubMed] [Google Scholar]
  • 18.Kamat PK, Vacek JC, Kalani A, Tyagi N. Homocysteine Induced Cerebrovascular Dysfunction: A Link to Alzheimer's Disease Etiology. The open neurology journal. 2015;9:9–14. doi: 10.2174/1874205X01509010009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Jansen WJ, Ossenkoppele R, Knol DL, Tijms BM, Scheltens P, Verhey FR, Visser PJ, Aalten P, Aarsland D, Alcolea D, Alexander M, Almdahl IS, Arnold SE, Baldeiras I, Barthel H, van Berckel BN, et al. Prevalence of cerebral amyloid pathology in persons without dementia: a meta-analysis. JAMA : the journal of the American Medical Association. 2015;313(19):1924–1938. doi: 10.1001/jama.2015.4668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Reitz C. Alzheimer's disease and the amyloid cascade hypothesis: a critical review. International journal of Alzheimer's disease. 2012;2012:369808. doi: 10.1155/2012/369808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Tang Z, Ioja E, Bereczki E, Hultenby K, Li C, Guan Z, Winblad B, Pei JJ. mTor mediates tau localization and secretion: implication for Alzheimer's disease. Biochimica et biophysica acta. 2015 doi: 10.1016/j.bbamcr.2015.03.003. [DOI] [PubMed] [Google Scholar]
  • 22.Zhu S, Shala A, Bezginov A, Sljoka A, Audette G, Wilson DJ. Hyperphosphorylation of intrinsically disordered tau protein induces an amyloidogenic shift in its conformational ensemble. PLoS One. 2015;10(3):e0120416. doi: 10.1371/journal.pone.0120416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Pimplikar SW. Reassessing the amyloid cascade hypothesis of Alzheimer's disease. The international journal of biochemistry & cell biology. 2009;41(6):1261–1268. doi: 10.1016/j.biocel.2008.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Herrup K. The case for rejecting the amyloid cascade hypothesis. Nature neuroscience. 2015;18(6):794–799. doi: 10.1038/nn.4017. [DOI] [PubMed] [Google Scholar]
  • 25.Rubio-Perez JM, Morillas-Ruiz JM. A review: inflammatory process in Alzheimer's disease, role of cytokines. TheScientificWorldJournal. 2012;2012:756357. doi: 10.1100/2012/756357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Doens D, Fernández PL. Microglia receptors and their implications in the response to amyloid beta for Alzheimer's disease pathogenesis. Journal of neuroinflammation. 2014;11:48. doi: 10.1186/1742-2094-11-48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.McGeer PL, McGeer EG. Innate immunity, local inflammation, and degenerative disease. Science of aging knowledge environment : SAGE KE. 2002;2002(29):re3. doi: 10.1126/sageke.2002.29.re3. [DOI] [PubMed] [Google Scholar]
  • 28.Heneka MT, Carson MJ, Khoury JE, Landreth GE, Brosseron F, Feinstein DL, Jacobs AH, Wyss-Coray T, Vitorica J, Ransohoff RM, Herrup K, Frautschy SA, Finsen B, Brown GC, Verkhratsky A, Yamanaka K, et al. Neuroinflammation in Alzheimer's disease. Lancet neurology. 2015;14(4):388–405. doi: 10.1016/S1474-4422(15)70016-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ong WY, Farooqui AA. Iron, neuroinflammation, and Alzheimer's disease. Journal of Alzheimer's disease : JAD. 2005;8(2):183–200. doi: 10.3233/jad-2005-8211. discussion 209-115. [DOI] [PubMed] [Google Scholar]
  • 30.Tuppo EE, Arias HR. The role of inflammation in Alzheimer's disease. The international journal of biochemistry & cell biology. 2005;37(2):289–305. doi: 10.1016/j.biocel.2004.07.009. [DOI] [PubMed] [Google Scholar]
  • 31.Tzeng SF, Hsiao HY, Mak OT. Prostaglandins and cyclooxygenases in glial cells during brain inflammation. Current drug targets Inflammation and allergy. 2005;4(3):335–340. doi: 10.2174/1568010054022051. [DOI] [PubMed] [Google Scholar]
  • 32.Varley J, Brooks DJ, Edison P. Imaging neuroinflammation in Alzheimer's and other dementias: Recent advances and future directions. Alzheimer's & dementia : the journal of the Alzheimer's Association. 2014 doi: 10.1016/j.jalz.2014.08.105. [DOI] [PubMed] [Google Scholar]
  • 33.Latta CH, Brothers HM, Wilcock DM. Neuroinflammation in Alzheimer's disease; A source of heterogeneity and target for personalized therapy. Neuroscience. 2014 doi: 10.1016/j.neuroscience.2014.09.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Castellani RJ, Moreira PI, Perry G, Zhu X. The role of iron as a mediator of oxidative stress in Alzheimer disease. BioFactors (Oxford, England) 2012;38(2):133–138. doi: 10.1002/biof.1010. [DOI] [PubMed] [Google Scholar]
  • 35.Connor JR, Menzies SL, St Martin SM, Mufson EJ. A histochemical study of iron, transferrin, and ferritin in Alzheimer's diseased brains. Journal of neuroscience research. 1992;31(1):75–83. doi: 10.1002/jnr.490310111. [DOI] [PubMed] [Google Scholar]
  • 36.Crichton RR, Dexter DT, Ward RJ. Brain iron metabolism and its perturbation in neurological diseases. Journal of neural transmission (Vienna, Austria : 1996) 2011;118(3):301–314. doi: 10.1007/s00702-010-0470-z. [DOI] [PubMed] [Google Scholar]
  • 37.Friedman A, Arosio P, Finazzi D, Koziorowski D, Galazka-Friedman J. Ferritin as an important player in neurodegeneration. Parkinsonism & related disorders. 2011;17(6):423–430. doi: 10.1016/j.parkreldis.2011.03.016. [DOI] [PubMed] [Google Scholar]
  • 38.Kell DB. Iron behaving badly: inappropriate iron chelation as a major contributor to the aetiology of vascular and other progressive inflammatory and degenerative diseases. BMC medical genomics. 2009;2:2. doi: 10.1186/1755-8794-2-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kell DB. Towards a unifying, systems biology understanding of large-scale cellular death and destruction caused by poorly liganded iron: Parkinson's, Huntington's, Alzheimer's, prions, bactericides, chemical toxicology and others as examples. Archives of toxicology. 2010;84(11):825–889. doi: 10.1007/s00204-010-0577-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Smith MA, Zhu X, Tabaton M, Liu G, McKeel DW, Jr, Cohen ML, Wang X, Siedlak SL, Dwyer BE, Hayashi T, Nakamura M, Nunomura A, Perry G. Increased iron and free radical generation in preclinical Alzheimer disease and mild cognitive impairment. Journal of Alzheimer's disease : JAD. 2010;19(1):363–372. doi: 10.3233/JAD-2010-1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wood H. Alzheimer disease: Iron-the missing link between ApoE and Alzheimer disease? Nature reviews Neurology. 2015;11(7):369. doi: 10.1038/nrneurol.2015.96. [DOI] [PubMed] [Google Scholar]
  • 42.Ayton S, Faux NG. Ferritin levels in the cerebrospinal fluid predict Alzheimer's disease outcomes and are regulated by APOE. 2015;6:6760. doi: 10.1038/ncomms7760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Yang H, Guan H, Yang M, Liu Z, Takeuchi S, Yanagisawa D, Vincent SR, Zhao S, Tooyama I. Upregulation of mitochondrial ferritin by proinflammatory cytokines: implications for a role in Alzheimer's disease. Journal of Alzheimer's disease : JAD. 2015;45(3):797–811. doi: 10.3233/JAD-142595. [DOI] [PubMed] [Google Scholar]
  • 44.Bandyopadhyay S, Rogers JT. Alzheimer's disease therapeutics targeted to the control of amyloid precursor protein translation: maintenance of brain iron homeostasis. Biochemical pharmacology. 2014;88(4):486–494. doi: 10.1016/j.bcp.2014.01.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Crespo ÂC, Silva B, Marques L, Marcelino E, Maruta C, Costa S, Timóteo A, Vilares A, Couto FS, Faustino P, Correia AP, Verdelho A, Porto G, Guerreiro M, Herrero A, Costa C, et al. Genetic and biochemical markers in patients with Alzheimer's disease support a concerted systemic iron homeostasis dysregulation. Neurobiology of aging. 2014;35(4):777–785. doi: 10.1016/j.neurobiolaging.2013.10.078. [DOI] [PubMed] [Google Scholar]
  • 46.Hadzhieva M, Kirches E, Mawrin C. Review: iron metabolism and the role of iron in neurodegenerative disorders. Neuropathology and applied neurobiology. 2014;40(3):240–257. doi: 10.1111/nan.12096. [DOI] [PubMed] [Google Scholar]
  • 47.Ward RJ, Dexter DT, Crichton RR. Neurodegenerative diseases and therapeutic strategies using iron chelators. Journal of trace elements in medicine and biology : organ of the Society for Minerals and Trace Elements (GMS) 2015;31:267–273. doi: 10.1016/j.jtemb.2014.12.012. [DOI] [PubMed] [Google Scholar]
  • 48.Collingwood JF, Davidson MR. The role of iron in neurodegenerative disorders: insights and opportunities with synchrotron light. Frontiers in pharmacology. 2014;5:191. doi: 10.3389/fphar.2014.00191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Casadesus G, Smith MA, Zhu X, Aliev G, Cash AD, Honda K, Petersen RB, Perry G. Alzheimer disease: evidence for a central pathogenic role of iron-mediated reactive oxygen species. Journal of Alzheimer's disease : JAD. 2004;6(2):165–169. doi: 10.3233/jad-2004-6208. [DOI] [PubMed] [Google Scholar]
  • 50.Crapper McLachlan DR, Dalton AJ, Kruck TP, Bell MY, Smith WL, Kalow W, Andrews DF. Intramuscular desferrioxamine in patients with Alzheimer's disease. Lancet. 1991;337(8753):1304–1308. doi: 10.1016/0140-6736(91)92978-b. [DOI] [PubMed] [Google Scholar]
  • 51.Pretorius E, Bester J, Vermeulen N, Lipinski B. Oxidation inhibits iron-induced blood coagulation. Current drug targets. 2013;14(1):13–19. doi: 10.2174/1389450111314010003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Pretorius E, Bester J, Vermeulen N, Lipinski B, Gericke GS, Kell DB. Profound morphological changes in the erythrocytes and fibrin networks of patients with hemochromatosis or with hyperferritinemia, and their normalization by iron chelators and other agents. PlosOne. 2014 doi: 10.1371/journal.pone.0085271. eCollection 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Pretorius E, Kell DB. Diagnostic morphology: biophysical indicators for iron-driven inflammatory diseases. Integrative Biology. 2014;6:486–510. doi: 10.1039/c4ib00025k. [DOI] [PubMed] [Google Scholar]
  • 54.Pretorius E, Lipinski B. Differences in Morphology of Fibrin Clots Induced with Thrombin and Ferric Ions and Its Pathophysiological Consequences. Heart, lung & circulation. 2012;22:447–449. doi: 10.1016/j.hlc.2012.10.010. [DOI] [PubMed] [Google Scholar]
  • 55.Pretorius E, Vermeulen N, Bester J, Lipinski B, Kell DB. A novel method for assessing the role of iron and its functional chelation in fibrin fibril formation: the use of scanning electron microscopy. Toxicology mechanisms and methods. 2013;23(5):352–359. doi: 10.3109/15376516.2012.762082. [DOI] [PubMed] [Google Scholar]
  • 56.Undas A, Ariëns RAS. Fibrin clot structure and function: a role in the pathophysiology of arterial and venous thromboembolic diseases. Arterioscler Thromb Vasc Biol. 2011;31(12):e88–99. doi: 10.1161/ATVBAHA.111.230631. [DOI] [PubMed] [Google Scholar]
  • 57.Kell DB, Pretorius E. Serum ferritin is an important inflammatory disease marker, as it is mainly a leakage product from damaged cells. Metallomics. 2014;4:748–773. doi: 10.1039/c3mt00347g. [DOI] [PubMed] [Google Scholar]
  • 58.Kell DB, Pretorius E. The simultaneous occurrence of both hypercoagulability and hypofibrinolysis in blood and serum during systemic inflammation, and the roles of iron and fibrin(ogen) Integrative Biology. 2015;7:24–52. doi: 10.1039/c4ib00173g. [DOI] [PubMed] [Google Scholar]
  • 59.Dávalos D, Akassoglou K. Fibrinogen as a key regulator of inflammation in disease. Semin Immunopathol. 2012;34(1):43–62. doi: 10.1007/s00281-011-0290-8. [DOI] [PubMed] [Google Scholar]
  • 60.Duvoix A, Dickens J, Haq I, Mannino D, Miller B, Tal-Singer R, Lomas DA. Blood fibrinogen as a biomarker of chronic obstructive pulmonary disease. Thorax. 2013;68(7):670–676. doi: 10.1136/thoraxjnl-2012-201871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Erikssen G, Liestol K, Bjornholt JV, Stormorken H, Thaulow E, Erikssen J. Erythrocyte sedimentation rate: a possible marker of atherosclerosis and a strong predictor of coronary heart disease mortality. European heart journal. 2000;21(19):1614–1620. doi: 10.1053/euhj.2000.2148. [DOI] [PubMed] [Google Scholar]
  • 62.Fries D, Martini WZ. Role of fibrinogen in trauma-induced coagulopathy. British journal of anaesthesia. 2010;105(2):116–121. doi: 10.1093/bja/aeq161. [DOI] [PubMed] [Google Scholar]
  • 63.Késmárky G, Kenyeres P, Késmárky M, Tóth K. Plasma viscosity: a forgotten variable. Clinical hemorheology and microcirculation. 2008;39(1-4):243–246. [PubMed] [Google Scholar]
  • 64.Kolodziejczyk J, Ponczek MB. The role of fibrinogen, fibrin and fibrin(ogen) degradation products (FDPs) in tumor progression. Contemporary oncology (Poznan, Poland) 2013;17(2):113–119. doi: 10.5114/wo.2013.34611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Pulanic D, Rudan I. The past decade: fibrinogen. Collegium antropologicum. 2005;29(1):341–349. [PubMed] [Google Scholar]
  • 66.Cortes-Canteli M, Paul J, Norris EH, Bronstein R, Ahn HJ, Zamolodchikov D, Bhuvanendran S, Fenz KM, Strickland S. Fibrinogen and beta-amyloid association alters thrombosis and fibrinolysis: a possible contributing factor to Alzheimer's disease. Neuron. 2010;66(5):695–709. doi: 10.1016/j.neuron.2010.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Narayan PJ, Kim SL, Lill C, Feng S, Faull RL, Curtis MA, Dragunow M. Assessing fibrinogen extravasation into Alzheimer's disease brain using high-content screening of brain tissue microarrays. Journal of neuroscience methods. 2015;247:41–49. doi: 10.1016/j.jneumeth.2015.03.017. [DOI] [PubMed] [Google Scholar]
  • 68.Cortes-Canteli M, Mattei L, Richards AT, Norris EH, Strickland S. Fibrin deposited in the Alzheimer's disease brain promotes neuronal degeneration. Neurobiology of aging. 2015;36(2):608–617. doi: 10.1016/j.neurobiolaging.2014.10.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Ahn HJ, Glickman JF, Poon KL, Zamolodchikov D, Jno-Charles OC, Norris EH, Strickland S. A novel Abeta-fibrinogen interaction inhibitor rescues altered thrombosis and cognitive decline in Alzheimer's disease mice. J Exp Med. 2014;211(6):1049–1062. doi: 10.1084/jem.20131751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Walton BL, Byrnes JR, Wolberg AS. Fibrinogen, red blood cells, and factor XIII in venous thrombosis. Journal of thrombosis and haemostasis : JTH. 2015;13:S208–215. doi: 10.1111/jth.12918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Ma X, Ji XM, Fu P, Ding YC, Xue Q, Huang Y. Coexistence of High Fibrinogen and Low High-density Lipoprotein Cholesterol Levels Predicts Recurrent Cerebral Venous Thrombosis. Chinese medical journal. 2015;128(13):1732–1737. doi: 10.4103/0366-6999.159345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Ahmed MS, Jadhav AB, Hassan A, Meng QH. Acute Phase Reactants as Novel Predictors of Cardiovascular Disease. ISRN inflammation. 2012;2012:953461. doi: 10.5402/2012/953461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ariëns RAS. Fibrin(ogen) and thrombotic disease. J Thromb Haemost. 2013;11:294–305. doi: 10.1111/jth.12229. [DOI] [PubMed] [Google Scholar]
  • 74.Barr JD, Chauhan AK, Schaeffer GV, Hansen JK, Motto DG. Red blood cells mediate the onset of thrombosis in the ferric chloride murine model. Blood. 2013;121(18):3733–3741. doi: 10.1182/blood-2012-11-468983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Day SM, Duquaine D, Mundada LV, Menon RG, Khan BV, Rajagopalan S, Fay WP. Chronic iron administration increases vascular oxidative stress and accelerates arterial thrombosis. Circulation. 2003;107(20):2601–2606. doi: 10.1161/01.CIR.0000066910.02844.D0. [DOI] [PubMed] [Google Scholar]
  • 76.Franchini M, Targher G, Montagnana M, Lippi G. Iron and thrombosis. Annals of hematology. 2008;87(3):167–173. doi: 10.1007/s00277-007-0416-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Lipinski B, Pretorius E. Iron-Induced Fibrin in Cardiovascular Disease. Current neurovascular research. 2013 doi: 10.2174/15672026113109990016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Lipinski B, Pretorius E, Oberholzer HM, van der Spuy WJ. Interaction of fibrin with red blood cells: the role of iron. Ultrastruct Pathol. 2012;36(2):79–84. doi: 10.3109/01913123.2011.627491. [DOI] [PubMed] [Google Scholar]
  • 79.Nielsen VG, Pretorius E. Iron-enhanced coagulation is attenuated by chelation A thrombelastographic and ultrastructural analysis. Blood coagulation & fibrinolysis : an international journal in haemostasis and thrombosis. 2014;25(8):845–850. doi: 10.1097/MBC.0000000000000160. [DOI] [PubMed] [Google Scholar]
  • 80.Singh N, Haldar S, Tripathi AK, McElwee MK, Horback K, Beserra A. Iron in Neurodegenerative Disorders of Protein Misfolding: A Case of Prion Disorders and Parkinson's Disease. Antioxidants & redox signaling. 2014 doi: 10.1089/ars.2014.5874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Pretorius E, Oberholzer HM, van der Spuy WJ, Meiring JH. Smoking and coagulation: the sticky fibrin phenomenon. Ultrastruct Pathol. 2010;34(4):236–239. doi: 10.3109/01913121003743716. [DOI] [PubMed] [Google Scholar]
  • 82.Pretorius E, Oberholzer HM, van der Spuy WJ, Swanepoel AC, Soma P. Scanning electron microscopy of fibrin networks in rheumatoid arthritis: a qualitative analysis. Rheumatology international. 2012;32(6):1611–1615. doi: 10.1007/s00296-011-1805-2. [DOI] [PubMed] [Google Scholar]
  • 83.Pretorius E, Swanepoel AC, Oberholzer HM, Van Der Spuy WJ, Duim W, Wessels PF. A descriptive investigation of the ultrastructure of fibrin networks in thrombo-embolic ischemic stroke. Journal of thrombosis and thrombolysis. 2011;31(4):507–513. doi: 10.1007/s11239-010-0538-5. [DOI] [PubMed] [Google Scholar]
  • 84.Pretorius E, Vermeulen N, Bester J, Lipinski B. Novel use of scanning electron microscopy for detection of iron-induced morphological changes in human blood. Microsc Res Tech. 2013;76(3):268–271. doi: 10.1002/jemt.22163. [DOI] [PubMed] [Google Scholar]
  • 85.Swanepoel AC, Nielsen VG, Pretorius E. Viscoelasticity and Ultrastructure in Coagulation and Inflammation: Two Diverse Techniques, One Conclusion. Inflammation eprint (ahead of print) 2015 doi: 10.1007/s10753-015-0148-7. [DOI] [PubMed] [Google Scholar]
  • 86.van Rooy M, Duim W, Ehlers R, Buys AV, Pretorius E. Platelet hyperactivity and fibrin clot structure in metabolic syndrome-induced transient ischemic attack: a microscopy and thromboelastography® study. Cardiovascular diabetology. 2015:Accepted. doi: 10.1186/s12933-015-0249-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Nielsen VG, Pretorius E. Iron and carbon monoxide enhance coagulation and attenuate fibrinolysis by different mechanisms. Blood coagulation & fibrinolysis : an international journal in haemostasis and thrombosis. 2014;25(7):695–702. doi: 10.1097/MBC.0000000000000128. [DOI] [PubMed] [Google Scholar]
  • 88.Nielsen VG, Pretorius E, Bester J, Jacobsen WK, Boyle PK, Reinhard JP. Carbon monoxide and iron modulate plasmatic coagulation in Alzheimer's disease. Current neurovascular research. 2015;12(1):31–39. doi: 10.2174/1567202612666150102150042. [DOI] [PubMed] [Google Scholar]
  • 89.Lipinski B, Pretorius E. Novel pathway of iron-induced blood coagulation: implications for diabetes mellitus and its complications. Polskie Archiwum Medycyny Wewnetrznej. 2012;122(3):115–122. [PubMed] [Google Scholar]
  • 90.Potgieter M, Bester J, Kell DB, Pretorius E. The dormant blood microbiome in chronic, inflammatory diseases. FEMS microbiology reviews. 2015;pii:fuv013. doi: 10.1093/femsre/fuv013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Kaprelyants AS, Gottschal JC, Kell DB. Dormancy in non-sporulating bacteria. FEMS microbiology reviews. 1993;10(3-4):271–285. doi: 10.1111/j.1574-6968.1993.tb05871.x. [DOI] [PubMed] [Google Scholar]
  • 92.Kell DB, Kaprelyants AS, Weichart DH, Harwood CR, Barer MR. Viability and activity in readily culturable bacteria: a review and discussion of the practical issues. Antonie van Leeuwenhoek. 1998;73(2):169–187. doi: 10.1023/a:1000664013047. [DOI] [PubMed] [Google Scholar]
  • 93.Shaik MM, Ahmad S, Gan SH, Abuzenadah AM, Ahmad E, Tabrez S, Ahmed F, Kamal MA. How do periodontal infections affect the onset and progression of Alzheimer's disease? CNS & neurological disorders drug targets. 2014;13(3):460–466. doi: 10.2174/18715273113126660152. [DOI] [PubMed] [Google Scholar]
  • 94.Honjo K, van Reekum R, Verhoeff NP. Alzheimer's disease and infection: do infectious agents contribute to progression of Alzheimer's disease? Alzheimer's & dementia : the journal of the Alzheimer's Association. 2009;5(4):348–360. doi: 10.1016/j.jalz.2008.12.001. [DOI] [PubMed] [Google Scholar]
  • 95.de Souza Rolim T, Fabri GM, Nitrini R, Anghinah R, Teixeira MJ, de Siqueira JT, Cestari JA, de Siqueira SR. Oral infections and orofacial pain in Alzheimer's disease: a case-control study. Journal of Alzheimer's disease : JAD. 2014;38(4):823–829. doi: 10.3233/JAD-131283. [DOI] [PubMed] [Google Scholar]
  • 96.Miklossy J. Alzheimer's disease - a neurospirochetosis. Analysis of the evidence following Koch's and Hill's criteria. Journal of neuroinflammation. 2011;8:90. doi: 10.1186/1742-2094-8-90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Itzhaki RF. Herpes simplex virus type 1 and Alzheimer's disease: increasing evidence for a major role of the virus. Frontiers in aging neuroscience. 2014;6:202. doi: 10.3389/fnagi.2014.00202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Hill JM, Bhattacharjee S, Pogue AI, Lukiw WJ. The gastrointestinal tract microbiome and potential link to Alzheimer's disease. Frontiers in neurology. 2014;5:43. doi: 10.3389/fneur.2014.00043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Hill JM, Clement C, Pogue AI, Bhattacharjee S, Zhao Y, Lukiw WJ. Pathogenic microbes, the microbiome, and Alzheimer's disease (AD) Frontiers in aging neuroscience. 2014;6:127. doi: 10.3389/fnagi.2014.00127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Hill JM, Lukiw WJ. Microbial-generated amyloids and Alzheimer's disease (AD) Frontiers in aging neuroscience. 2015;7:9. doi: 10.3389/fnagi.2015.00009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Kamer AR, Dasanayake AP, Craig RG, Glodzik-Sobanska L, Bry M, de Leon MJ. Alzheimer's disease and peripheral infections: the possible contribution from periodontal infections, model and hypothesis. Journal of Alzheimer's disease : JAD. 2008;13(4):437–449. doi: 10.3233/jad-2008-13408. [DOI] [PubMed] [Google Scholar]
  • 102.Gaur S, Agnihotri R. Alzheimer's disease and chronic periodontitis: Is there an association? Geriatrics & gerontology international. 2014 doi: 10.1111/ggi.12425. [DOI] [PubMed] [Google Scholar]
  • 103.Farhad SZ, Amini S, Khalilian A, Barekatain M, Mafi M, Barekatain M, Rafei E. The effect of chronic periodontitis on serum levels of tumor necrosis factor-alpha in Alzheimer disease. Dental research journal. 2014;11(5):549–552. [PMC free article] [PubMed] [Google Scholar]
  • 104.Sparks Stein P, Steffen MJ, Smith C, Jicha G, Ebersole JL, Abner E, Dawson D., 3rd Serum antibodies to periodontal pathogens are a risk factor for Alzheimer's disease. Alzheimer's & dementia : the journal of the Alzheimer's Association. 2012;8(3):196–203. doi: 10.1016/j.jalz.2011.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Kamer AR, Craig RG, Dasanayake AP, Brys M, Glodzik-Sobanska L, de Leon MJ. Inflammation and Alzheimer's disease: possible role of periodontal diseases. Alzheimer's & dementia : the journal of the Alzheimer's Association. 2008;4(4):242–250. doi: 10.1016/j.jalz.2007.08.004. [DOI] [PubMed] [Google Scholar]
  • 106.Singhrao SK, Harding A, Simmons T, Robinson S, Kesavalu L, Crean S. Oral inflammation, tooth loss, risk factors, and association with progression of Alzheimer's disease. Journal of Alzheimer's disease : JAD. 2014;42(3):723–737. doi: 10.3233/JAD-140387. [DOI] [PubMed] [Google Scholar]
  • 107.Poole S, Singhrao SK, Kesavalu L, Curtis MA, Crean S. Determining the presence of periodontopathic virulence factors in short-term postmortem Alzheimer's disease brain tissue. Journal of Alzheimer's disease : JAD. 2013;36(4):665–677. doi: 10.3233/JAD-121918. [DOI] [PubMed] [Google Scholar]
  • 108.Miklossy J. Historic evidence to support a causal relationship between spirochetal infections and Alzheimer's disease. Frontiers in aging neuroscience. 2015;7:46. doi: 10.3389/fnagi.2015.00046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Ransmayr GN, Holliger S, Schletterer K, Heidler H, Deibl M, Poewe W, Madersbacher H, Kiss G. Lower urinary tract symptoms in dementia with Lewy bodies, Parkinson disease, and Alzheimer disease. Neurology. 2008;70(4):299–303. doi: 10.1212/01.wnl.0000296826.61499.26. [DOI] [PubMed] [Google Scholar]
  • 110.Eriksson I, Gustafson Y, Fagerstrom L, Olofsson B. Urinary tract infection in very old women is associated with delirium. International psychogeriatrics / IPA. 2011;23(3):496–502. doi: 10.1017/S1041610210001456. [DOI] [PubMed] [Google Scholar]
  • 111.Ogama N, Yoshida M, Nakai T, Niida S, Toba K, Sakurai T. Frontal white matter hyperintensity predicts lower urinary tract dysfunction in older adults with amnestic mild cognitive impairment and Alzheimer's disease. Geriatrics & gerontology international. 2015 doi: 10.1111/ggi.12447. [DOI] [PubMed] [Google Scholar]
  • 112.Asti A, Gioglio L. Can a bacterial endotoxin be a key factor in the kinetics of amyloid fibril formation? Journal of Alzheimer's disease : JAD. 2014;39(1):169–179. doi: 10.3233/JAD-131394. [DOI] [PubMed] [Google Scholar]
  • 113.Kanekiyo T, Xu H, Bu G. ApoE and Abeta in Alzheimer's disease: accidental encounters or partners? Neuron. 2014;81(4):740–754. doi: 10.1016/j.neuron.2014.01.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Schmidt V, Carlo AS, Willnow TE. Apolipoprotein E receptor pathways in Alzheimer disease. Wiley interdisciplinary reviews Systems biology and medicine. 2014;6(3):255–270. doi: 10.1002/wsbm.1262. [DOI] [PubMed] [Google Scholar]
  • 115.Wang C, Yu JT, Wang HF, Jiang T, Tan CC, Meng XF, Soares HD, Tan L. Meta-analysis of peripheral blood apolipoprotein E levels in Alzheimer's disease. PLoS One. 2014;9(2):e89041. doi: 10.1371/journal.pone.0089041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Šerý O, Hlinecká L, Balcar VJ, Janout V, Povova J. Diabetes, hypertension and stroke - does Alzheimer protect you? Neuro endocrinology letters. 2015;35(8):691–696. [PubMed] [Google Scholar]
  • 117.Rodriguez GA, Tai LM, LaDu MJ, Rebeck GW. Human APOE4 increases microglia reactivity at Abeta plaques in a mouse model of Abeta deposition. Journal of neuroinflammation. 2014;11:111. doi: 10.1186/1742-2094-11-111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Saura J, Petegnief V, Wu X, Liang Y, Paul SM. Microglial apolipoprotein E and astroglial apolipoprotein J expression in vitro: opposite effects of lipopolysaccharide. Journal of neurochemistry. 2003;85(6):1455–1467. doi: 10.1046/j.1471-4159.2003.01788.x. [DOI] [PubMed] [Google Scholar]
  • 119.Cunningham C, Wilcockson DC, Campion S, Lunnon K, Perry VH. Central and systemic endotoxin challenges exacerbate the local inflammatory response and increase neuronal death during chronic neurodegeneration. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2005;25(40):9275–9284. doi: 10.1523/JNEUROSCI.2614-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Lee JW, Lee YK, Yuk DY, Choi DY, Ban SB, Oh KW, Hong JT. Neuro-inflammation induced by lipopolysaccharide causes cognitive impairment through enhancement of beta-amyloid generation. Journal of neuroinflammation. 2008;5:37. doi: 10.1186/1742-2094-5-37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Herrera AJ, Tomás-Camardiel M, Venero JL, Cano J, Machado A. Inflammatory process as a determinant factor for the degeneration of substantia nigra dopaminergic neurons. Journal of neural transmission (Vienna, Austria : 1996) 2005;112(1):111–119. doi: 10.1007/s00702-004-0121-3. [DOI] [PubMed] [Google Scholar]
  • 122.De Pablos RM, Herrera AJ, Villaran RF, Cano J, Machado A. Dopamine-dependent neurotoxicity of lipopolysaccharide in substantia nigra. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2005;19(3):407–409. doi: 10.1096/fj.04-2153fje. [DOI] [PubMed] [Google Scholar]
  • 123.Byler SL, Boehm GW, Karp JD, Kohman RA, Tarr AJ, Schallert T, Barth TM. Systemic lipopolysaccharide plus MPTP as a model of dopamine loss and gait instability in C57Bl/6J mice. Behav Brain Res. 2009;198(2):434–439. doi: 10.1016/j.bbr.2008.11.027. [DOI] [PubMed] [Google Scholar]
  • 124.Kelly LP, Carvey PM, Keshavarzian A, Shannon KM, Shaikh M, Bakay RA, Kordower JH. Progression of intestinal permeability changes and alpha-synuclein expression in a mouse model of Parkinson's disease. Movement disorders : official journal of the Movement Disorder Society. 2014;29(8):999–1009. doi: 10.1002/mds.25736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Hoban DB, Connaughton E, Connaughton C, Hogan G, Thornton C, Mulcahy P, Moloney TC, Dowd E. Further characterisation of the LPS model of Parkinson's disease: a comparison of intra-nigral and intra-striatal lipopolysaccharide administration on motor function, microgliosis and nigrostriatal neurodegeneration in the rat. Brain, behavior, and immunity. 2013;27(1):91–100. doi: 10.1016/j.bbi.2012.10.001. [DOI] [PubMed] [Google Scholar]
  • 126.Hritcu L, Ciobica A. Intranigral lipopolysaccharide administration induced behavioral deficits and oxidative stress damage in laboratory rats: relevance for Parkinson's disease. Behav Brain Res. 2013;253:25–31. doi: 10.1016/j.bbr.2013.07.006. [DOI] [PubMed] [Google Scholar]
  • 127.Hritcu L, Ciobica A, Stefan M, Mihasan M, Palamiuc L, Nabeshima T. Spatial memory deficits and oxidative stress damage following exposure to lipopolysaccharide in a rodent model of Parkinson's disease. Neuroscience research. 2011;71(1):35–43. doi: 10.1016/j.neures.2011.05.016. [DOI] [PubMed] [Google Scholar]
  • 128.Tufekci KU, Genc S, Genc K. The endotoxin-induced neuroinflammation model of Parkinson's disease. Parkinson's disease. 2011;2011:487450. doi: 10.4061/2011/487450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Zhan X, Cox C, Ander BP, Liu D, Stamova B, Jin LW, Jickling GC, Sharp FR. Inflammation Combined with Ischemia Produces Myelin Injury and Plaque-Like Aggregates of Myelin, Amyloid-beta and AbetaPP in Adult Rat Brain. Journal of Alzheimer's disease : JAD. 2015 doi: 10.3233/JAD-143072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Deng X, Li M, Ai W, He L, Lu D, Patrylo PR, Cai H, Luo X, Li Z, Yan X. Lipolysaccharide-Induced Neuroinflammation Is Associated with Alzheimer-Like Amyloidogenic Axonal Pathology and Dendritic Degeneration in Rats. Advances in Alzheimer's disease. 2014;3(2):78–93. doi: 10.4236/aad.2014.32009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Xu Z, Wu J, Zheng J, Ma H, Zhang H, Zhen X, Zheng LT, Zhang X. Design, synthesis and evaluation of a series of non-steroidal anti-inflammatory drug conjugates as novel neuroinflammatory inhibitors. International immunopharmacology. 2015;25(2):528–537. doi: 10.1016/j.intimp.2015.02.033. [DOI] [PubMed] [Google Scholar]
  • 132.de Punder K, Pruimboom L. Stress induces endotoxemia and low-grade inflammation by increasing barrier permeability. Frontiers in immunology. 2015;6:223. doi: 10.3389/fimmu.2015.00223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Zhang XY, Cao JB, Zhang LM, Li YF, Mi WD. Deferoxamine attenuates lipopolysaccharide-induced neuroinflammation and memory impairment in mice. Journal of neuroinflammation. 2015;12(1):20. doi: 10.1186/s12974-015-0238-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Daulatzai MA. Role of stress, depression, and aging in cognitive decline and Alzheimer's disease. Current topics in behavioral neurosciences. 2014;18:265–296. doi: 10.1007/7854_2014_350. [DOI] [PubMed] [Google Scholar]
  • 135.Jain NK, Patil CS, Kulkarni SK, Singh A. Modulatory role of cyclooxygenase inhibitors in aging- and scopolamine or lipopolysaccharide-induced cognitive dysfunction in mice. Behav Brain Res. 2002;133(2):369–376. doi: 10.1016/s0166-4328(02)00025-6. [DOI] [PubMed] [Google Scholar]
  • 136.Davis DH, Skelly DT, Murray C, Hennessy E, Bowen J, Norton S, Brayne C, Rahkonen T, Sulkava R, Sanderson DJ, Rawlins JN, Bannerman DM, MacLullich AM, Cunningham C. Worsening cognitive impairment and neurodegenerative pathology progressively increase risk for delirium. The American journal of geriatric psychiatry : official journal of the American Association for Geriatric Psychiatry. 2015;23(4):403–415. doi: 10.1016/j.jagp.2014.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Chu AJ, Rauci M, Nwobi OI, Mathews ST, Beydoun S. Novel anticoagulant activity of polyamino acid offsets bacterial endotoxin-induced extrinsic hypercoagulation: downregulation of monocytic tissue factor-dependent FVII activation. Journal of cardiovascular pharmacology. 2003;42(4):477–483. doi: 10.1097/00005344-200310000-00004. [DOI] [PubMed] [Google Scholar]
  • 138.Chu AJ, Wang ZG, Raicu M, Beydoun S, Ramos N. Protamine inhibits tissue factor-initiated extrinsic coagulation. British journal of haematology. 2001;115(2):392–399. doi: 10.1046/j.1365-2141.2001.03100.x. [DOI] [PubMed] [Google Scholar]
  • 139.Slotta JE, Braun OO, Menger MD, Thorlacius H. Central role of rho kinase in lipopolysaccharide-induced platelet capture on venous endothelium. Journal of investigative medicine : the official publication of the American Federation for Clinical Research. 2008;56(4):720–725. doi: 10.2310/JIM.0b013e31816c3e81. [DOI] [PubMed] [Google Scholar]
  • 140.Gorog DA, Jeong YH. Platelet function tests: why they fail to guide personalized antithrombotic medication. Journal of the American Heart Association. 2015;4(5) doi: 10.1161/JAHA.115.002094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Yamamoto J, Inoue N, Otsui K, Ishii H, Gorog DA. Global Thrombosis Test (GTT) can detect major determinants of haemostasis including platelet reactivity, endogenous fibrinolytic and thrombin generating potential. Thrombosis research. 2014;133(5):919–926. doi: 10.1016/j.thromres.2014.02.018. [DOI] [PubMed] [Google Scholar]
  • 142.Ghanim H, Abuaysheh S, Sia CL, Korzeniewski K, Chaudhuri A, Fernandez-Real JM, Dandona P. Increase in plasma endotoxin concentrations and the expression of Toll-like receptors and suppressor of cytokine signaling-3 in mononuclear cells after a high-fat, high-carbohydrate meal: implications for insulin resistance. Diabetes care. 2009;32(12):2281–2287. doi: 10.2337/dc09-0979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Opal SM, Scannon PJ, Vincent JL, White M, Carroll SF, Palardy JE, Parejo NA, Pribble JP, Lemke JH. Relationship between plasma levels of lipopolysaccharide (LPS) and LPS-binding protein in patients with severe sepsis and septic shock. The Journal of infectious diseases. 1999;180(5):1584–1589. doi: 10.1086/315093. [DOI] [PubMed] [Google Scholar]
  • 144.Zweigner J, Gramm HJ, Singer OC, Wegscheider K, Schumann RR. High concentrations of lipopolysaccharide-binding protein in serum of patients with severe sepsis or septic shock inhibit the lipopolysaccharide response in human monocytes. Blood. 2001;98(13):3800–3808. doi: 10.1182/blood.v98.13.3800. [DOI] [PubMed] [Google Scholar]
  • 145.Leante-Castellanos JL, de Guadiana-Romualdo LG, Fuentes-Gutierrez C, Hernando-Holgado A, Garcia-Gonzalez A, Jimenez-Santos E. The value of lipopolysaccharide binding protein for diagnosis of late-onset neonatal sepsis in very low birth weight infants. Journal of perinatal medicine. 2015;43(2):253–257. doi: 10.1515/jpm-2014-0010. [DOI] [PubMed] [Google Scholar]
  • 146.Funderburg NT, Stubblefield Park SR, Sung HC, Hardy G, Clagett B, Ignatz-Hoover J, Harding CV, Fu P, Katz JA, Lederman MM, Levine AD. Circulating CD4(+) and CD8(+) T cells are activated in inflammatory bowel disease and are associated with plasma markers of inflammation. Immunology. 2013;140(1):87–97. doi: 10.1111/imm.12114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Broadhurst D, Kell DB. Statistical strategies for avoiding false discoveries in metabolomics and related experiments. Metabolomics. 2006;2:171–196. [Google Scholar]
  • 148.Leek JT, Peng RD. Statistics: P values are just the tip of the iceberg. Nature. 2015;520(7549):612. doi: 10.1038/520612a. [DOI] [PubMed] [Google Scholar]
  • 149.Nielsen VG, Kirklin H.K., Hoogendoorn H, Ellis T.C., Holman W.L. Thromboelastographic method to quantify the contribution of factor XIII to coagulation kinetics. Blood coagulation & Fibrinolysis: an international journal in haemostasis and thrombosis. 2007;18(2):145–150. doi: 10.1097/MBC.0b013e32802f7d91. [DOI] [PubMed] [Google Scholar]
  • 150.Nielsen VG. Beyond cell based models of coagulation: analyses of coagulation with clot “lifespan” resistance-time relationships. Thrombosis research. 2008;122(2):145–152. doi: 10.1016/j.thromres.2007.09.003. [DOI] [PubMed] [Google Scholar]
  • 151.Nielsen VG, Gurley WQ, Burch TM. The Impact of Factor XIII on Coagulation Kinetics and Clot Strength Determined by Thrombelastography. Anesthesia & Analgesia. 2004;99(1):120–123. doi: 10.1213/01.ANE.0000123012.24871.62. [DOI] [PubMed] [Google Scholar]
  • 152.Nielsen VG, Lyerly RT, Gurley WQ. The effect of dilution on plasma coagulation kinetics determined by thrombelastography is dependent on antithrombin activity and mode of activation. Anesthesia and analgesia. 2004;99(6):1587–1592. doi: 10.1213/01.ANE.0000136843.58799.AB. [DOI] [PubMed] [Google Scholar]
  • 153.Haemonetics . TEG 5000 Hemostasis analyzer system. Massachusetts: Haemonetics Corporation; 2011. [Google Scholar]
  • 154.Nielsen VG, Audu P, Cankovic L, Lyerly RT, 3rd, Steenwyk BL, Armstead V, Powell G. Qualitative thrombelastographic detection of tissue factor in human plasma. Anesthesia and analgesia. 2007;104(1):59–64. doi: 10.1213/01.ane.0000248223.05152.a1. [DOI] [PubMed] [Google Scholar]
  • 155.Buys AV, Van Rooy MJ, Soma P, Van Papendorp D, Lipinski B, Pretorius E. Changes in red blood cell membrane structure in type 2 diabetes: a scanning electron and atomic force microscopy study. Cardiovascular diabetology. 2013;12(1):25. doi: 10.1186/1475-2840-12-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Saraf S, Christopoulos C, Salha IB, Stott DJ, Gorog DA. Impaired endogenous thrombolysis in acute coronary syndrome patients predicts cardiovascular death and nonfatal myocardial infarction. Journal of the American College of Cardiology. 2010;55(19):2107–2115. doi: 10.1016/j.jacc.2010.01.033. [DOI] [PubMed] [Google Scholar]
  • 157.Hare DJ, Doecke JD, Faux NG, Rembach A, Volitakis I, Fowler CJ, Grimm R, Doble PA, Cherny RA, Masters CL, Bush AI, Roberts BR. Decreased plasma iron in Alzheimer's disease is due to transferrin desaturation. ACS chemical neuroscience. 2015;6(3):398–402. doi: 10.1021/cn5003557. [DOI] [PubMed] [Google Scholar]
  • 158.Brandhagen DJ, Fairbanks VF, Baldus W. Recognition and management of hereditary hemochromatosis. American family physician. 2002;65(5):853–860. [PubMed] [Google Scholar]
  • 159.Barton JC, McDonnell SM, Adams PC, Brissot P, Powell LW, Edwards CQ, Cook JD, Kowdley KV. Management of hemochromatosis. Hemochromatosis Management Working Group. Annals of internal medicine. 1998;129(11):932–939. doi: 10.7326/0003-4819-129-11_part_2-199812011-00003. [DOI] [PubMed] [Google Scholar]
  • 160.Bester J, Buys AV, Lipinski B, Kell DB, Pretorius E. High ferritin levels have major effects on the morphology of erythrocytes in Alzheimer's disease. Frontiers in aging neuroscience. 2013 doi: 10.3389/fnagi.2013.00088. eCollection 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Ritchie RF, Palomaki GE, Neveux LM, Navolotskaia O, Ledue TB, Craig WY. Reference distributions for the negative acute-phase serum proteins, albumin, transferrin and transthyretin: a practical, simple and clinically relevant approach in a large cohort. Journal of clinical laboratory analysis. 1999;13(6):273–279. doi: 10.1002/(SICI)1098-2825(1999)13:6&#x0003c;273::AID-JCLA4&#x0003e;3.0.CO;2-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Giometto B, Argentiero V, Sanson F, Ongaro G, Tavolato B. Acute-phase proteins in Alzheimer's disease. European neurology. 1988;28(1):30–33. doi: 10.1159/000116224. [DOI] [PubMed] [Google Scholar]
  • 163.Pretorius E, Swanepoel AC, Buys AV, Vermeulen N, Duim W, Kell DB. Eryptosis as a marker of Parkinson's disease. Aging-US. 2014;6:788–818. doi: 10.18632/aging.100695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Pretorius E, Vermeulen N, Bester J, du Plooy JL, Gericke GS. The effect of iron overload on red blood cell morphology. The Lancet. 2014;383:722. doi: 10.1016/S0140-6736(13)61208-8. [DOI] [PubMed] [Google Scholar]
  • 165.Pretorius E. The adaptability of red blood cells. Cardiovascular diabetology. 2013;12:63. doi: 10.1186/1475-2840-12-63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.van Rooy M, Pretorius E. Metabolic syndrome, platelet activation and the development of transient ischemic attack or thromboembolic stroke. Thrombosis research. 2015;135(3):434–442. doi: 10.1016/j.thromres.2014.12.030. [DOI] [PubMed] [Google Scholar]
  • 167.Lang F, Qadri SM. Mechanisms and significance of eryptosis, the suicidal death of erythrocytes. Blood purification. 2012;33(1-3):125–130. doi: 10.1159/000334163. [DOI] [PubMed] [Google Scholar]
  • 168.Lang E, Qadri SM, Lang F. Killing me softly - suicidal erythrocyte death. The international journal of biochemistry & cell biology. 2012;44(8):1236–1243. doi: 10.1016/j.biocel.2012.04.019. [DOI] [PubMed] [Google Scholar]
  • 169.Qadri SM, Kucherenko Y, Zelenak C, Jilani K, Lang E, Lang F. Dicoumarol activates Ca2+-permeable cation channels triggering erythrocyte cell membrane scrambling. Cellular physiology and biochemistry : international journal of experimental cellular physiology, biochemistry, and pharmacology. 2011;28(5):857–864. doi: 10.1159/000335800. [DOI] [PubMed] [Google Scholar]
  • 170.Qadri SM, Bauer J, Zelenak C, Mahmud H, Kucherenko Y, Lee SH, Ferlinz K, Lang F. Sphingosine but not sphingosine-1-phosphate stimulates suicidal erythrocyte death. Cellular physiology and biochemistry : international journal of experimental cellular physiology, biochemistry, and pharmacology. 2011;28(2):339–346. doi: 10.1159/000331750. [DOI] [PubMed] [Google Scholar]
  • 171.Pretorius E, Oberholzer HM, van der Spuy WJ, Swanepoel AC, Soma P. Qualitative scanning electron microscopy analysis of fibrin networks and platelet abnormalities in diabetes. Blood coagulation & fibrinolysis : an international journal in haemostasis and thrombosis. 2011;22(6):463–467. doi: 10.1097/MBC.0b013e3283468a0d. [DOI] [PubMed] [Google Scholar]
  • 172.Van Der Spuy WJ, Pretorius E. A place for ultrastructural analysis of platelets in cerebral ischemic research. Microsc Res Tech. 2013;76(8):795–802. doi: 10.1002/jemt.22231. [DOI] [PubMed] [Google Scholar]
  • 173.Ai Q, Jing Y, Jiang R, Lin L, Dai J, Che Q, Zhou D, Jia M, Wan J, Zhang L. Rotenone, a mitochondrial respiratory complex I inhibitor, ameliorates lipopolysaccharide/D-galactosamine-induced fulminant hepatitis in mice. International immunopharmacology. 2014;21(1):200–207. doi: 10.1016/j.intimp.2014.04.028. [DOI] [PubMed] [Google Scholar]
  • 174.Alpsoy S, Akyuz A, Erfan G, Akkoyun DC, Topcu B, Guzel S, Kaya S, Kulac M. Atherosclerosis, some serum inflammatory markers in psoriasis. Giornale italiano di dermatologia e venereologia : organo ufficiale, Societa italiana di dermatologia e sifilografia. 2014;149(2):167–175. [PubMed] [Google Scholar]
  • 175.Amar J, Lange C, Payros G, Garret C, Chabo C, Lantieri O, Courtney M, Marre M, Charles MA, Balkau B, Burcelin R, Group DESIRS Blood microbiota dysbiosis is associated with the onset of cardiovascular events in a large general population: the D.E.S.I.R. study. PLoS One. 2013;8(1):e54461. doi: 10.1371/journal.pone.0054461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Aksu G, Ozturk C, Kavakli K, Genel F, Kutukculer N. Hypercoagulability: interaction between inflammation and coagulation in familial Mediterranean fever. Clinical rheumatology. 2007;26(3):366–370. doi: 10.1007/s10067-006-0334-y. [DOI] [PubMed] [Google Scholar]
  • 177.Anžej S, Božič M, Antovič A, Peternel P, Gašperšič N, Rot U, Tratar G, Stegnar M. Evidence of hypercoagulability and inflammation in young patients long after acute cerebral ischaemia. Thrombosis research. 2007;120(1):39–46. doi: 10.1016/j.thromres.2006.08.005. [DOI] [PubMed] [Google Scholar]
  • 178.Arshad F, Lisman T, Porte RJ. Hypercoagulability as a contributor to thrombotic complications in the liver transplant recipient. Liver international : official journal of the International Association for the Study of the Liver. 2013;33(6):820–827. doi: 10.1111/liv.12140. [DOI] [PubMed] [Google Scholar]
  • 179.Choi G, Schultz MJ, Levi M, van der Poll T. The relationship between inflammation and the coagulation system. Swiss Med Wkly. 2006;136(9-10):139–144. doi: 10.4414/smw.2006.11059. [DOI] [PubMed] [Google Scholar]
  • 180.Cicala C, Cirino G. Linkage between inflammation and coagulation: an update on the molecular basis of the crosstalk. Life sciences. 1998;62(20):1817–1824. doi: 10.1016/s0024-3205(97)01167-3. [DOI] [PubMed] [Google Scholar]
  • 181.Conway EM. Thrombomodulin and its role in inflammation. Semin Immunopathol. 2012;34(1):107–125. doi: 10.1007/s00281-011-0282-8. [DOI] [PubMed] [Google Scholar]
  • 182.Emsley HC, Tyrrell PJ. Inflammation and infection in clinical stroke. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism. 2002;22(12):1399–1419. doi: 10.1097/01.WCB.0000037880.62590.28. [DOI] [PubMed] [Google Scholar]
  • 183.Esmon CT. The interactions between inflammation and coagulation. British journal of haematology. 2005;131(4):417–430. doi: 10.1111/j.1365-2141.2005.05753.x. [DOI] [PubMed] [Google Scholar]
  • 184.Koch L, Hofer S, Weigand MA, Frommhold D, Poeschl J. Lipopolysaccharide-induced activation of coagulation in neonatal cord and adult blood monitored by thrombelastography. Thrombosis research. 2009;124(4):463–467. doi: 10.1016/j.thromres.2009.05.002. [DOI] [PubMed] [Google Scholar]

Articles from Oncotarget are provided here courtesy of Impact Journals, LLC

RESOURCES