Platelets, the small, anucleate blood cells that originate from megakaryocytes in the bone marrow, are typically associated with coagulation. However, it is now apparent that platelets are more multifaceted than originally thought, with their function extending beyond their traditional role in hemostasis to acting as important mediators of brain function. In this review, we outline the broad repertoire of platelet function in the central nervous system, focusing on the similarities between platelets and neurons. We also summarize the role that platelets play in the pathophysiology of various neurological diseases, with a particular focus on neuroinflammation and neurodegeneration. Finally, we highlight the exciting prospect of harnessing the unique features of the platelet proteome and extracellular vesicles, which are rich in neurotrophic, antioxidative, and antiinflammatory factors, for the development of novel neuroprotective and neuroregenerative interventions to treat various neurodegenerative and traumatic pathologies.

Platelets, the smallest blood cells, are derived from megakaryocytes and are most well known for their role in clot formation. However, less than one-tenth of the circulating platelets are required to facilitate hemostasis, suggesting that these cells likely fulfill additional functions.1-4 Platelets are now recognized as essential components of the immune system, capable of participating in both innate and adaptive immune responses.1-4 They contribute to innate immunity through their ability to release a plethora of inflammatory molecules upon activation, which attract or modulate effector cells and direct leukocytes toward sites of inflammation. Platelets also express Toll-like receptors, which can directly bind pathogens via the detection of pathogen-associated molecule patterns.5 In addition, growing evidence suggests that they mediate adaptive immune responses via their expression of functional CD40L.6 Platelets can also deliver antigens to antigen-presenting cells,7 process and present antigens themselves,8 and influence T-cell responses.9 

Despite containing no nucleus, and therefore playing no role in transcription, platelets contain messenger RNA (mRNA), a functional proteasome containing antigen-presentation machinery,10 and a surprisingly complete set of organelles and storage granules, including mitochondria, lysosomes, α (α)- and dense granules rich in small molecules and trophic factors, a dense tubular system analogous to the endoplasmic reticulum, and a highly invaginated plasma membrane system known as the open canalicular system (Figure 1). The human platelet proteome is currently estimated to contain approximately 5200 proteins.11 Although a recent study revealed a high similarity between the transcriptomes of platelets and megakaryocytes, many transcripts are not expressed in platelets, suggesting a redistribution of RNA species inherited from megakaryocytes following platelet shedding in the blood.11 There is, however, a good relationship between the platelet transcriptome and proteome, indicating the capacity for platelets to carry out translational processes.11 In addition, the open canalicular system, which makes up approximately 1% of the platelet volume, is filled with proteins captured from the plasma by diffusion and endocytosis.

Figure 1.

Major features and biological cargo of platelets. Platelets contain various organelles and storage granules, including mitochondria, lysosomes, α- and dense granules, a dense tubular system, and a highly invaginated plasma membrane system known as the open canalicular system. The α- and dense granules and lysosomes contain a plethora of small molecules and trophic factors that are released upon activation.

Figure 1.

Major features and biological cargo of platelets. Platelets contain various organelles and storage granules, including mitochondria, lysosomes, α- and dense granules, a dense tubular system, and a highly invaginated plasma membrane system known as the open canalicular system. The α- and dense granules and lysosomes contain a plethora of small molecules and trophic factors that are released upon activation.

Close modal

The largest and most abundant of the platelet granules are the α-granules. Between 200 and 500 nm in diameter and numbering approximately 50 to 80 per cell, these granules resemble the large dense-core vesicles of neurons and contain proteins such as growth factors, cytokines, antioxidants, and adhesion molecules that are either synthesized in the megakaryocytes before platelet segmentation or taken up from the plasma by exocytosis. The dense granules are smaller (200-300 nm in diameter), approximately 10-fold fewer in number than α-granules, and resemble the small dense-core vesicles of neurons. These granules primarily contain small molecules such as serotonin and other neurotransmitters, adenosine triphosphate, adenosine diphosphate (ADP), and calcium, which are taken up from extracellular pools. Platelets also typically have 1 to 2 lysosomes per cell that contain acid hydrolases, cathepsins, and lysosomal-associated membrane glycoproteins.

To fulfill particular functions, platelets must first be activated. Distinct subsets of proteins are secreted in response to specific activation stimuli, as evidenced by their characteristic response to 3 common agonists: ADP, collagen, and thrombin receptor-activating peptide.12 In addition, the existence of subpopulations of α-granules, in which unique subsets of proteins are stored and released by stimulation with specific agonists, further highlights the finely tuned mechanism by which bioactive molecules are released by platelets in a context-dependent manner.13 In addition to the direct secretion of bioactive molecules from granules, platelets also release extracellular vesicles (EVs).14 First described as “platelet dust” in the 1960s,15 EVs are emerging as important mediators of intercellular communication. The majority of EVs in the blood are platelet- or megakaryocyte-derived.16,17 Although resting platelets constitutively produce EVs,18 activation increases their number and defines their specific cargo.19 Platelets are believed to predominantly release 2 types of EVs: platelet microvesicles (PMVs) and exosomes.14 PMVs (0.1-1 µm in diameter) are shed from the plasma membrane20 and, like platelets, carry selective subsets of RNA, proteins, and organelles such as mitochondria, with their heterogeneity being dictated by the activation stimulus which triggers their formation.21,22 However, the precise mechanism by which they selectively package and release their biological cargo remains unclear. Exosomes (30-100 nm in diameter) are smaller than PMVs and are derived from endosomal multivesicular bodies and α-granules.23 Interestingly, activated platelets also release respiratory-competent mitochondria as free organelles that can act as damage-associated molecular patterns in several pathologies.24 

In addition to their role in the periphery, it is becoming increasingly clear that platelets also contribute to both the degeneration and regeneration of the central nervous system (CNS). Both platelets and the EVs they release are small enough to traverse the microcapillaries spanning the brain under both healthy and pathological conditions. This enables crosstalk with neural cells via a number of mechanisms, either indirectly via the secretion of bioactive molecules from platelet granules, exosomes, or PMVs, or directly by local receptor-mediated interactions.25 Interestingly, platelets also exhibit a remarkable number of similarities to neurons, further highlighting the potential for crosstalk between these cells and the brain.

Despite their different embryonic origins, with neurons arising from the ectoderm and platelets from megakaryocytes that originate from the mesoderm, it has been proposed that, in many ways, the communication between platelets and other immune cells, including neutrophils, monocytes, and clusters of differentiation (CD)8+ T cells, mirrors the interaction between pre- and postsynaptic neuronal terminals26-28 (Figure 2). These similarities may facilitate the crosstalk between platelets and neurons in the wide range of neurodegenerative disorders in which platelet dysfunction has been implicated. In a mechanism analogous to that which occurs at the presynapse of neurons, calcium-dependent activation of the platelet secretory machinery triggers the fusion of internal vesicles with the plasma membrane via the specific, highly conserved docking molecules, SNAREs (soluble NSF attachment protein receptors), resulting in the rapid vesicular release of neurotransmitters.29 There is, however, a marked difference in the kinetics of signal transduction and exocytosis between platelets and neurons, with neurotransmitter release occurring 10 000 times faster in neurons.29,30 Although increases in intracellular Ca2+ trigger exocytosis in both cell types, the magnitude and mechanism of the calcium signal generation differ significantly. In neurons, membrane depolarization results in Ca2+ influx via Ca2+ channels, which leads to a local Ca2+ increase of ∼200 mmol/L and triggers the exocytosis of nearby vesicles. In contrast, platelet granule secretion is mediated by the activation of extracellular receptors, which increases the concentration of intracellular cytosolic Ca2+ (to ∼10 mmol/L) and activates protein kinase C. Moreover, unlike platelets which typically undergo irreversible aggregation following activation, neurons rapidly return to their resting state and can be reactivated following a refractory period of approximately 1 ms.

Figure 2.

Platelets and neurons share many similarities. The top panel shows the communication between a presynaptic and a postsynaptic neuron. Neurons are activated by calcium signaling, which triggers the release of neurotransmitters such as serotonin, dopamine, epinephrine, histamine, glutamate, and GABA. The neurotransmitters then bind to the corresponding receptors on the postsynaptic neuron to complete neurotransmission. The lower panel shows an activated platelet with dense granules containing neurotransmitters, adenosine triphosphate, adenosine diphosphate, and calcium, and α-granules containing growth factors, cytokines, antioxidants, and adhesion molecules. Similar to the mechanism which occurs at the presynapse of neurons, calcium-dependent activation of platelets triggers the fusion of the granules with the plasma membrane via highly conserved docking molecules called SNAREs. This results in the rapid release of neurotransmitters, which bind to corresponding receptors expressed on the surface of T cells.

Figure 2.

Platelets and neurons share many similarities. The top panel shows the communication between a presynaptic and a postsynaptic neuron. Neurons are activated by calcium signaling, which triggers the release of neurotransmitters such as serotonin, dopamine, epinephrine, histamine, glutamate, and GABA. The neurotransmitters then bind to the corresponding receptors on the postsynaptic neuron to complete neurotransmission. The lower panel shows an activated platelet with dense granules containing neurotransmitters, adenosine triphosphate, adenosine diphosphate, and calcium, and α-granules containing growth factors, cytokines, antioxidants, and adhesion molecules. Similar to the mechanism which occurs at the presynapse of neurons, calcium-dependent activation of platelets triggers the fusion of the granules with the plasma membrane via highly conserved docking molecules called SNAREs. This results in the rapid release of neurotransmitters, which bind to corresponding receptors expressed on the surface of T cells.

Close modal

Platelets also carry a plethora of neurotransmitters crucial for intercellular communication between neurons, together with the receptors and transporters of these molecules, further highlighting the crosstalk and homology between the periphery and the CNS. The most abundant of these is the monoamine serotonin.31 Platelets take up serotonin from the plasma via the serotonin transporter and store it in dense granules. The release of serotonin from dense granules is a marker of the degree of platelet activation while also contributing to platelet aggregation, the recruitment of circulating platelets, and the promotion of coagulation. In addition to serotonin, platelets carry several other neurotransmitters crucial for the intercellular communication between neurons, including other biogenic amines such as epinephrine, dopamine, and histamine, the excitatory neurotransmitter glutamate, and the inhibitory neurotransmitter γ-aminobutyric acid (GABA). Glutamate is the most abundant excitatory neurotransmitter in the brain, and as in neurons, substrate-induced uptake of glutamate in human platelets most likely occurs via glutamate receptors.32 Platelets store significant amounts of GABA, although the concentrations are approximately 30% lower than in neurons.33 

Platelets also transport unusually high concentrations of several other proteins with important neuronal functions. The protein reelin, which regulates cell migration and synaptic plasticity, is carried by platelets, where it modifies platelet cytoskeletal dynamics and contributes to arterial thrombosis.34,35 Following platelet activation, reelin is secreted and binds to the amyloid precursor protein (APP) and apolipoprotein E receptor 2 to induce signaling.35 Interestingly, brain-derived neurotrophic factor (BDNF), a secretory protein that is primarily known for its role in regulating the development and function of neural circuits, accumulates in human platelets at 100- to 1000-fold higher levels than in neurons.36 During hemostasis, BDNF is released by platelets and modulates thrombosis.37,38 Surprisingly, however, BDNF is absent in mouse platelets and serum,39 suggesting that its biological role in platelets may be compensated by other platelet-derived growth factors. Other well-characterized platelet trophic factors that are important in mediating brain function include growth factors (eg, platelet-derived growth factor-AA and -BB, transforming growth factor-β, epidermal growth factor, vascular endothelial growth factor, and basic fibroblast growth factor), chemokines (eg, CXCL4 [platelet factor 4], CCL5 [chemokine ligand 5], and CCL3), and various antioxidants such as glutathione peroxidase.40-42 However, despite the similarities between neurons and platelets, why these cells have evolved to resemble each other remains unclear.

Neural plasticity refers to the ability of neural networks within the brain to change through growth or reorganization in response to experience. One way in which the brain retains lifelong plasticity is via the continuous addition of newly generated neurons into the existing circuitry, a process known as adult neurogenesis. The neurogenic niches are highly vascularized, with the neural stem cells lying in direct contact with blood vessels.43-45 It has even been suggested that platelets should be considered as part of the neurovascular unit,46 which typically includes neurons, glial cells, neural precursor cells (NPCs), endothelial cells, vascular smooth muscle cells, and pericytes.

Several studies have shown that platelets can modulate NPC proliferation and neurogenesis in the subventricular zone (SVZ), although this has only been studied in vitro or in the context of brain injury. Hayon and colleagues were the first to demonstrate that platelet microparticles can increase embryonic NPC proliferation, survival, and neuronal-lineage differentiation potential in vitro.47 Another study found that platelet lysate increased the number of in vitro proliferating adult rat SVZ-derived NPCs by enhancing their survival via a reduction in apoptosis without affecting their proliferative or lineage-differentiation capacity.46 Neurogenesis-enhancing effects of platelets have also been observed in animal models of stroke.46,48,49 Recently, platelets were found to regulate in vitro hippocampal neurogenesis, with activated platelets increasing the proliferation and neuronal lineage differentiation potential of the primary dentate gyrus-derived NPCs.50 Physical exercise is one of the strongest physiological mediators of hippocampal neurogenesis. Platelets were found to contribute to a running-induced increase in mouse hippocampal neurogenesis, with induction of thrombocytopenia abolishing the exercise-induced increase in NPC proliferation, indicating that platelets are required for this response.50 However, no difference in baseline neurogenesis amounts was observed between the sedentary platelet-depleted and control mice. Interestingly, a proteomic comparison revealed that none of the classical platelet activation markers, such as the activation-specific glycoprotein (GP) IIb and IIIa receptor,51 was differentially expressed following exercise, suggesting that exercise-induced platelet activation is a distinct process.50 

Another way in which the brain retains plasticity is via changes that occur at synapses, the junctions that allow for communication between neurons. Neurovascular damage, such as traumatic brain injury (TBI), and neurovascular diseases, such as stroke, typically result in neuroinflammation accompanied by an increase in neuronal plasticity. Dukhinova and colleagues52 proposed that dynamic platelet–neuron synapse-like structures form after brain injury, which induces platelets to release serotonin,53 thereby stimulating neurite growth and the formation of new synapses.52 Platelets were also found to upregulate the expression of several genes involved in neuronal synaptic plasticity, stimulating the formation of dendritic spines and new synapses.52,54 In support of these findings, an earlier study found that platelet-activating factor can induce hippocampal long-term potentiation, a form of activity-driven synaptic plasticity that involves a persistent strengthening of synapses, leading to a long-lasting increase in signal transmission between neurons.55 

In the previous sections, we discussed the physiological links between neurons and platelets. In the second half of this review, we will shift our focus to disease processes. Neuroinflammation, which contributes to the pathophysiology of various neurological diseases, comprises a cascade of accumulating damage encompassing cellular and molecular components, including the generation of reactive oxygen species, the release of cytokines and chemokines, microglial and astrocytic activation, neuronal protein unfolding and aggregation, and neuronal cell death.56,57 It is now becoming clear that systemic inflammation, neurovascular dysfunction, and coagulopathy coexist in neurological pathologies, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and multiple sclerosis (MS),58,59 highlighting a likely role of the blood compartment in the detrimental inflammatory aspect of neurological diseases. The concept that the blood system is involved in the pathophysiology of brain neuroinflammation is supported by the high level of vascularization of the CNS parenchyma, which facilitates crosstalk between the blood and the brain, and the loss of integrity of the blood–brain barrier (BBB) that occurs in various chronic neurodegenerative disorders and as a consequence of brain trauma.60 Among the multiple cellular and protein components of blood, platelets are thought to provide a link between the blood and the brain in disease,25,61 as evidenced by platelet invasion and deposition in brain lesions.59 

Platelets play a critical role in inflammatory processes and immune responses62 but also exhibit antiinflammatory potential through the regulation of macrophage function and regulatory T cells and the secretion of antiinflammatory factors.63 The functional capacity of platelets to act as inflammatory mediators (and hence to potentially modulate neuroinflammation) is supported by the fact that they are the first cells to reach sites of injury or inflammation. Platelets are therefore highly reactive to discrete changes that affect the plasma coagulation and complement systems, leading to their activation, exposure of functional membrane receptors,64 degranulation, and the release of trophic factors, cytokines, and chemokines.64 Under inflammatory states, platelets also release EVs that act as complementary proinflammatory mediators, capable of infiltrating tissues65 and capturing and activating neutrophils.66 

Various components expressed by activated platelets and platelet EVs can promote leukocyte–endothelial interactions and trigger inflammatory processes.66 Those components include phosphatidylserine, a coagulant phospholipid, a range of surface glycoproteins, proinflammatory cytokines and chemokines, as well as matrix metalloproteinases.59,67 In line with this, a recent study found that the activated platelet-mediated release of pathogenic serotonin after exposure to systemic immune complexes is a critical mediator of shock.68 

The interaction between platelets and the CNS involves crosstalk between activated platelets and vascular endothelial cell receptors and the recruitment of leukocytes and neutrophils from the blood circulation. Strikingly, activated platelets have been shown to affect the BBB integrity and infiltrate the brain69 in conditions such as stroke70 and MS,71 and interact with the cellular components of the BBB matrix (endothelial cells, pericytes, perivascular microglia, and astrocytes).72 Soluble CD40L, for example, was found to be a key contributor to astrocytic and microglial activation in hypertension, which increases the expression of adhesion molecules on the brain endothelium and results in deposition of platelets in the brain, leading to neuroinflammation and neuronal injury.73 Platelet–neuron associations have also been observed in the hippocampal parenchyma in animal models of MS, an observation with potential therapeutic relevance for treating the neuropsychiatric symptoms of the disease.71 Moreover, direct contact with brain glycolipid structures present in the membranes of astrocytes further activates platelets and stimulates neuroinflammation,53 leading to immune cell recruitment into the brain parenchyma74 and the simultaneous release of trophic factors.27,53,72 Interestingly, this cascade of events creates a microenvironment where proinflammatory cytokines, neurotrophic factors, and neurotransmitters coexist, which, in response to CNS damage, can stimulate new synapse formation and axonal regrowth.27 A specific role in the modulation of neuroinflammation has been assigned to the α-granule chemoattractants CXCL4 and CCL5 through monocyte recruitment, or MIP-1a (macrophage inflammatory protein-1a)67,75 because of interaction with immune cells, BBB cellular components, astrocytes and microglia, as well as neurons themselves. Together, these observations highlight the contribution of various platelet factors in neuroinflammation and have provided incentives for preclinical studies where the protective effect of antiplatelet adjunct therapy, such as aspirin, has been examined.76 The potential of various new classes of oral antiplatelet agents is being evaluated in preclinical and clinical studies, most notably to diminish ischemic stroke recurrence, albeit the risk of increasing intracranial bleeding needs to be carefully considered. These agents include vorapaxar, which blocks PAR-1 (protease-activated receptor-1); clopidogrel and ticagrelor, which inhibit P2Y12; revacept, a GPVI (glycoprotein VI) fusion protein; ACT101, a monoclonal anti-GPVI antibody; caplacizumab, an anti-von Willebrand factor nanobody; and cilostazol, a PDE-3 (phosphodiesterase-3) inhibitor of platelet aggregation. Another antiplatelet agent, ibudilast, which inhibits PDE-4, has been shown to slow brain atrophy in MS patients, consistent with the increased detrimental prothrombotic effect of platelets. Further studies are, however, required to delineate the therapeutic role of antiplatelet therapies in neurological disorders.75 

It is clear that neurodegenerative diseases not only involve the cells within the CNS, but that systemic mediators such as platelets also play an important role in their manifestation, with platelet dysfunction being observed in disorders including AD, PD, Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS), and MS. As such, platelets have been described as “circulating mirrors of neurons,”77 providing an easily accessible peripheral biomarker to monitor the onset and progression of these conditions. The key alterations in the physical parameters and molecular pathways of platelets in these conditions are summarized in Table 1.

Table 1.

Key platelet abnormalities linked to neurodegenerative conditions

ConditionKey platelet abnormalitiesSpecies/modelReferences
AD Increased platelet activation Human 134  
 Impaired platelet degranulation Human 135  
 Platelet hyperactivity APP23 mice 96  
 Increased platelet adhesion to subendothelial matrix components 3xTg-AD mice 95  
 Increased platelet invasion into brain parenchyma APP/PS1 mice 72  
 Platelet inclusions in cerebral blood vessels APP_SweDI mice 136  
 Decreased platelet mitochondrial respiration rate Human 137  
 Platelet mitochondrial cytochrome oxidase deficiency Human 138  
 Reduced platelet mitochondrial pyruvate dehydrogenase & respiratory complex IV Human 139  
 Increased platelet β-secretase activity Human 140  
 Increased activity of τ Human 104,106,141 
 Increased secretion of amyloid β Human 93  
 Altered ratio of APP isoforms Human 103  
 Increased glycogen synthase kinase 3-β Human 142  
 Reduced glutamate uptake Human 143  
PD Increased mean platelet volume Human 144  
 Decreased platelet glutamate uptake Human 145  
 Reduction in platelet vesicular monoamine transporter 2 mRNA Human 146  
 Platelet mitochondrial dysfunction Human 147,148 
 Increased platelet monoamine oxidase B MPTP mouse model 149  
HD Increased platelet mHtt protein levels Human 150  
 Increased platelet aspartate and glycine levels Human 151  
 Impaired platelet adenosine A receptor signaling Human 152  
 Impaired platelet nitric oxide metabolism Human 153  
 Elevated platelet mitochondrial monoamine oxidase activity Human 141  
ALS Altered platelet mitochondrial membrane potential Human 154  
 Altered platelet mitochondrial morphology Human 155  
 Increased platelet TDP-43 levels Human 156  
 Reduced complex IV activity in platelet mitochondria Human 157  
 Decreased platelet serotonin levels Human 158  
 Increased glutamate synthase resulting in decreased glutamate uptake Human 159  
 Increased thrombospondin Human 160  
MS Increased platelet activation Human 161  
 Increased platelet adhesiveness Human 162  
 Increased PMP release Human 163  
 Altered serotonin release from dense granules Human/EAE mice 164  
 Altered lipid composition of platelet membranes (decreased cholesterol linoleate) Human 165  
ConditionKey platelet abnormalitiesSpecies/modelReferences
AD Increased platelet activation Human 134  
 Impaired platelet degranulation Human 135  
 Platelet hyperactivity APP23 mice 96  
 Increased platelet adhesion to subendothelial matrix components 3xTg-AD mice 95  
 Increased platelet invasion into brain parenchyma APP/PS1 mice 72  
 Platelet inclusions in cerebral blood vessels APP_SweDI mice 136  
 Decreased platelet mitochondrial respiration rate Human 137  
 Platelet mitochondrial cytochrome oxidase deficiency Human 138  
 Reduced platelet mitochondrial pyruvate dehydrogenase & respiratory complex IV Human 139  
 Increased platelet β-secretase activity Human 140  
 Increased activity of τ Human 104,106,141 
 Increased secretion of amyloid β Human 93  
 Altered ratio of APP isoforms Human 103  
 Increased glycogen synthase kinase 3-β Human 142  
 Reduced glutamate uptake Human 143  
PD Increased mean platelet volume Human 144  
 Decreased platelet glutamate uptake Human 145  
 Reduction in platelet vesicular monoamine transporter 2 mRNA Human 146  
 Platelet mitochondrial dysfunction Human 147,148 
 Increased platelet monoamine oxidase B MPTP mouse model 149  
HD Increased platelet mHtt protein levels Human 150  
 Increased platelet aspartate and glycine levels Human 151  
 Impaired platelet adenosine A receptor signaling Human 152  
 Impaired platelet nitric oxide metabolism Human 153  
 Elevated platelet mitochondrial monoamine oxidase activity Human 141  
ALS Altered platelet mitochondrial membrane potential Human 154  
 Altered platelet mitochondrial morphology Human 155  
 Increased platelet TDP-43 levels Human 156  
 Reduced complex IV activity in platelet mitochondria Human 157  
 Decreased platelet serotonin levels Human 158  
 Increased glutamate synthase resulting in decreased glutamate uptake Human 159  
 Increased thrombospondin Human 160  
MS Increased platelet activation Human 161  
 Increased platelet adhesiveness Human 162  
 Increased PMP release Human 163  
 Altered serotonin release from dense granules Human/EAE mice 164  
 Altered lipid composition of platelet membranes (decreased cholesterol linoleate) Human 165  

EAE, experimental autoimmune encephalomyelitis; mHtt, mutant huntingtin protein; PMP, platelet microparticle; TDP-43, TAR DNA-binding protein of 43 kDa.

The links between platelet dysfunction and neurodegeneration have been reviewed in detail elsewhere.61,77-80 Therefore, in the following section, we have chosen to highlight the links between platelets and AD, an age-related neurodegenerative disorder and the most prevalent form of dementia. The pathological hallmarks of this disease are extracellular deposits of amyloid-β (Aβ), intraneuronal aggregates of hyperphosphorylated τ, activated microglia, reactive astrocytes, dystrophic neurites, inflammation, and oxidative stress. Several studies have linked both normal platelet function and platelet dysfunction to the development of AD. Platelets are the primary source of circulating APP from which they can produce and subsequently secrete Aβ.81,82 Outside the CNS, platelets are the main source of Aβ.83,84 Both Aβ and all 3 isoforms of APP are expressed in megakaryocytes and stored in platelet α-granules from which they are released upon activation. Whereas neurons produce significant amounts of the Aβ42 peptide, platelets mainly produce Aβ40, which they release upon activation, contributing to approximately 90% of the Aβ in the blood.82,85 Unlike neurons, which predominantly process APP via the β-secretase pathway, platelets primarily process APP and release Aβ through α-secretase via the regulated secretory vesicle pathway.86,87 Nonetheless, platelets contain all 3 secretases and, in principle, could therefore generate all APP cleavage products.88 The physiological role of APP and its cleavage products in platelets remains unclear, although the antimicrobial effect of Aβ is clearly established, suggesting that it may act as a normal component of the innate immune system.89,90 Circulating Aβ can also cross the BBB.91 It was recently shown that platelets accelerate the amyloidosis process by disrupting the permeability of the BBB and accelerating the accumulation of Aβ in the brain parenchyma, thereby leading to the manifestation of AD in animal models.92-94 Together, a number of studies indicate that, in AD, platelets become hyperactive and release and interact with Aβ to accelerate disease progression,92,95,96 suggesting that the disease may, at least in part, be a slowly developing thrombohemorrhagic disorder.97 

Parabiosis between transgenic AD and wild-type mice has been shown to increase Aβ in the vessel walls and brain parenchyma and drive the formation of AD-like protein aggregates in the brain of the latter animals.98 Similarly, another study found that plasma from young mice reduced both Aβ and τ pathologies and neuroinflammation and ameliorated cognitive impairment in transgenic AD mice.99 In contrast, a study using human APP transgenic mice found that exposure to young blood did not alter amyloid plaque deposition.100 Although clinical trials in which AD patients receive young plasma are currently underway,101,102 the contribution of platelets to the properties and composition of this young plasma remains unknown.

In addition to the role of platelet-derived Aβ in AD pathogenesis, platelets may also serve as a peripheral biomarker. The ratio of platelet APP isoforms is significantly altered in AD patients, enabling the detection of conversion from mild cognitive impairment to AD.103 Although less well-studied, τ is also present in platelets. The ratio of high molecular weight forms to low molecular weight monomeric τ was found to be significantly higher in the platelets of AD patients.104,105 More recently, the levels of platelet Ser396/404 phosphorylated τ and Thr231 phosphorylated τ were found to be significantly higher in mild cognitive impairment patients and could be used to distinguish between them and normal subjects.106 

Despite considerable research, it remains unclear whether platelet dysfunction occurs before or because of the neurodegenerative process. Nevertheless, dampening platelet activation may represent a therapeutic approach to AD. Several studies have investigated antiplatelet agents as potential therapeutic compounds in AD. Of these, the most well-studied is low-dose aspirin, which has been shown to suppress the secretion of APP from human platelets.107 Unfortunately, however, the majority of studies have indicated that it is not effective in preventing or treating the disease.108,109 In contrast, the effects of another antiplatelet agent, cilostazol, appear to be more positive.110-112 Further studies are therefore required to fully elucidate the role of platelet activation in AD pathophysiology in order to develop new diagnostic and prognostic biomarkers as well as therapeutic approaches for the disease.

In addition to the neurodegenerative diseases discussed above, platelet dysfunction has been implicated in the neurological manifestations of other conditions. Immune thrombocytopenia is associated with neurological complications following many viral infections, including hepatitis C virus, human immunodeficiency virus, cytomegalovirus, Epstein-Barr virus, hantavirus, herpes virus, severe acute respiratory syndrome coronavirus, dengue virus, and Zika virus, potentially via platelet-induced BBB breakdown.113 Likewise, platelets have been shown to contribute to BBB breakdown in cerebral malaria, resulting in severe neurological complications.114 It has also been proposed that the increased platelet activation observed in diabetic patients contributes to an increased risk of developing AD via the release of inflammatory factors and Aβ.115 Moreover, essential thrombocythemia, an acquired myeloproliferative neoplasm that leads to elevated platelet concentrations, is associated with usually transient neurological symptoms, including blurred vision, headache, tinnitus, and dizziness. Although antiplatelet therapy is often used to treat these conditions, further research is required to fully elucidate the effectiveness of this approach.

Healthy platelet granules contain an amazing reservoir of potent antiinflammatory, neuroprotective, and antioxidative molecules63 that can potentially be harnessed for the treatment of neurodegenerative conditions. The translational rationale for using platelets in this way is linked not only to their high content of potent biomolecules but also to the fact that clinical platelet concentrates are readily available in many countries, thereby facilitating access to this potential biotherapy.116 In addition, the efficacy of platelet concentrates that are pathogen-reduced but no longer suitable for direct transfusion for cell therapy and regenerative medicine has been demonstrated in cell culture and animal models.116,117 Processing of platelets as source material of human platelet lysate (HPL) biologicals also benefits from the technical and regulatory experience gained with other blood-derived medicinal products.118 

Recent preclinical work has evaluated human platelet-rich plasma or platelet concentrates as source materials for neurotrophin-rich platelet lysates and EV preparations for the treatment of CNS diseases40,49,117,119-125 (Table 2). For example, the administration of HPL to the lateral ventricles of rats subjected to focal ischemia induced by permanent distal middle cerebral artery occlusion significantly enhanced the proliferation of endogenous neural stem cells and angiogenesis in the SVZ and in the perilesion cortex and improved neurobehavioral outcomes.49 Similarly, in an AD model, intranasal administration of HPL exerted a neuroprotective action, which led to decreased Aβ deposition, τ phosphorylation, and astrocyte reactivity, thereby preventing synaptic loss and stimulating global improvements in anxiety, learning, and memory behaviors.120 Intranasal delivery of HPL also decreased neuroinflammation and improved motor performance in a mouse model of PD.122 Consistent with these data, the intranasal administration of purified HPL resulted in robust protection of the dopaminergic neurons in the substantia nigra and striatum.119 This platelet lysate diffused to various brain areas, including the olfactory bulb, striatum, and cortex, without detectable induction of neuroinflammation, suggesting that its intranasal delivery may offer therapeutic potential against several neurodegenerative diseases.119 The multifaceted therapeutic potential of HPL has also recently been confirmed in models of TBI.40 The HPL was first applied topically in the lesioned area to mimic a severe TBI in which there would be brain access, followed by daily intranasal administration for 6 days after injury. This treatment significantly improved motor function, downregulated proinflammatory genes, controlled the level of reactive oxygen species, and reduced the loss of cortical synaptic proteins.

Table 2.

Preclinical studies of human platelet lysate and extracellular vesicle preparations for the treatment of neurological diseases

MaterialDiseasePlatelet preparation, model, and administration routeBiological effectsReferences
HPL Stroke PMCAO Intracerebroventricular NSC proliferation and angiogenesis Improved cognition 49  
 AD Primary cortical and hippocampal neurons APP/PS1 mouse model; intranasal delivery Proliferation and survival of primary neurons Hippocampal neurogenesis and reduced Aβ-induced neurodegeneration 121  
 PD Human dopaminergic neuroblastoma cell line (SH-SY5Y) treated with MPP+MPTP mouse model; intranasal administration Protection against MPP+ toxicity Diminished inflammatory responses Improved motor performance 122  
  LUHMES cells exposed to MPP+MPTP mouse model; intranasal administration In vitro and in vivo protection of dopaminergic neurons 119  
  LUHMES cells exposed to prooxidants and regulated cell death inducers Strong neuroprotection through activation of the Akt and MEK pathways 123  
  LUHMES cells; primary cortical and hippocampal neurons Enhanced expression of tyrosine hydroxylase and neuron-specific enolase in LUHMES cells and strong protection against ferroptosis 117  
 BI and TBI Proliferating SEZ-derived NPCs Increased proliferating SEZ-derived NPCs maintaining proliferative or lineage-differentiation capacity 46  
  Undifferentiated SH-SY5Y neuroblastoma cells; Ea-hy926 human endothelial cell; BV2 microglial cells No toxicity and no inflammation Stimulation of wound healing and neuronal differentiation of SH-SY5Y 125  
  Scratch assay (differentiated SH-SY5Y cells) Mouse models of TBI; topical followed by daily intranasal administration Wound healing Improved motor function; mitigation of cortical neuroinflammation and oxidative stress in the injured area; reduced loss of cortical synaptic proteins 40  
 ALS NSC34 motoneurons exposed to various neurotoxins Strong neuroprotection through activation of the Akt and MEK pathways 123  
  Motoneuron cultures SOD1G86R; intracerebroventricular or intranasal administration Akt-dependent neuroprotection, antiapoptotic and antiferroptotic actions Increased lifespan 126  
EVs Stroke PMCAO in vivo model; topical application by biodegradable polymer Neurogenesis and angiogenesis Improved behavioral function 48  
 BI and TBI In vitro assay of NSC proliferation, survival, and differentiation Increased proliferation, survival, and differentiation, through ERK and Akt signaling 47  
  Wound healing (SH-SY5Y) Mouse primary neuronal cells Nontoxicity Promotion of cell growth and migration Stimulation of network formation in primary neuronal cultures 130  
MaterialDiseasePlatelet preparation, model, and administration routeBiological effectsReferences
HPL Stroke PMCAO Intracerebroventricular NSC proliferation and angiogenesis Improved cognition 49  
 AD Primary cortical and hippocampal neurons APP/PS1 mouse model; intranasal delivery Proliferation and survival of primary neurons Hippocampal neurogenesis and reduced Aβ-induced neurodegeneration 121  
 PD Human dopaminergic neuroblastoma cell line (SH-SY5Y) treated with MPP+MPTP mouse model; intranasal administration Protection against MPP+ toxicity Diminished inflammatory responses Improved motor performance 122  
  LUHMES cells exposed to MPP+MPTP mouse model; intranasal administration In vitro and in vivo protection of dopaminergic neurons 119  
  LUHMES cells exposed to prooxidants and regulated cell death inducers Strong neuroprotection through activation of the Akt and MEK pathways 123  
  LUHMES cells; primary cortical and hippocampal neurons Enhanced expression of tyrosine hydroxylase and neuron-specific enolase in LUHMES cells and strong protection against ferroptosis 117  
 BI and TBI Proliferating SEZ-derived NPCs Increased proliferating SEZ-derived NPCs maintaining proliferative or lineage-differentiation capacity 46  
  Undifferentiated SH-SY5Y neuroblastoma cells; Ea-hy926 human endothelial cell; BV2 microglial cells No toxicity and no inflammation Stimulation of wound healing and neuronal differentiation of SH-SY5Y 125  
  Scratch assay (differentiated SH-SY5Y cells) Mouse models of TBI; topical followed by daily intranasal administration Wound healing Improved motor function; mitigation of cortical neuroinflammation and oxidative stress in the injured area; reduced loss of cortical synaptic proteins 40  
 ALS NSC34 motoneurons exposed to various neurotoxins Strong neuroprotection through activation of the Akt and MEK pathways 123  
  Motoneuron cultures SOD1G86R; intracerebroventricular or intranasal administration Akt-dependent neuroprotection, antiapoptotic and antiferroptotic actions Increased lifespan 126  
EVs Stroke PMCAO in vivo model; topical application by biodegradable polymer Neurogenesis and angiogenesis Improved behavioral function 48  
 BI and TBI In vitro assay of NSC proliferation, survival, and differentiation Increased proliferation, survival, and differentiation, through ERK and Akt signaling 47  
  Wound healing (SH-SY5Y) Mouse primary neuronal cells Nontoxicity Promotion of cell growth and migration Stimulation of network formation in primary neuronal cultures 130  

BI, brain injury; NSC, neural stem cell; PMCAO, permanent distal middle cerebral artery occlusion; SEZ, subependymal zone.

The use of platelet lysates or purified fractions to treat ALS has also been investigated.124 Platelet lysate treatment of NSC-34 (a spinal cord and neuroblastoma hybrid cell line) motor neurons exposed to staurosporine and menadione toxins resulted in strong neuroprotection.123 A HPL and its fractions separated based on molecular mass also induced protein kinase B (also known as Akt)-dependent neuroprotection as well as a strong antiapoptotic and antiferroptotic action in neuronal cell cultures.126 The low molecular weight (<3 kDa) fraction had glutathione peroxidase 4-dependent antiferroptotic properties, suggesting a protective role of platelet factors other than the higher molecular weight neurotrophins. Consistent with the in vitro findings, the lifespan of superoxide dismutase 1G86R mice, a model of ALS, was significantly increased by intracerebroventricular delivery of the complete lysate and intranasal administration of the <3 kDa fraction,126 expanding the scope of potential therapeutic applications.

There is also increasing interest in therapeutic applications of platelet EVs as drug delivery systems due to their ability to transport trophic factors, cross tissue barriers, diffuse into tissues, and target cells. The possibility of using EVs, rather than free molecules or nanocarriers,127 as neurotrophic factor-rich therapeutic formulations against CNS disorders has been evidenced using mesenchymal stem cell EVs in preclinical models of neurological diseases.128 Clinical-grade platelet concentrates and HPL contain as many as 1012 EVs/mL,129,130 with a mean diameter of 50 to 300 nm and a slightly negative ζ potential that can be exploited for brain diffusion and the delivery of neurotrophic factors.130 Similar to platelets, EVs carry growth factors, cytokines, and chemokines, as well as antiinflammatory molecules and microRNAs,131 although whether they play a critical role in the neuroprotective function of HPL remains unclear, as does their impact on HPL safety for brain administration.131,132 However, compared with unfractionated HPLs, EV administration may decrease protein overload in the CNS, prolong protection and the release of trophic factors, and support the targeting of pathological brain tissues.133 Preliminary evidence has revealed that EVs purified from various HPLs are nontoxic and capable of stimulating neuroblastoma cell healing as well as favoring network formation in primary cortical neuronal cultures.130 It has also been found that platelet EVs promote the in vitro proliferation and differentiation of neural stem cells47 and functionally stimulate neurogenesis when topically applied with a biodegradable polymer in an in vivo stroke model, further demonstrating the ability of EVs to diffuse within the brain.48 Together, these data support the need for further evaluation of platelet EVs as a standalone biotherapy or drug delivery vehicle in neuroregenerative medicine.

As summarized above, there is now considerable evidence that the functions of platelets extend well beyond their canonical role in hemostasis. Platelet dysfunction has been implicated in several neurodegenerative and neuroinflammatory conditions, opening fascinating new perspectives for a deeper understanding of the pathophysiology of these disorders. However, whether platelet dysfunction initiates the onset of pathophysiology or arises as a consequence of it remains unclear. Nevertheless, knowledge of the crosstalk between platelets and the brain parenchyma will likely drive the development of novel antiplatelet therapies for the prevention or management of CNS disorders. Healthy platelet-derived preparations are also gaining increasing recognition as a potential neuroprotective biotherapy. Although further preclinical and clinical investigation is required, the possibility of administering well-characterized and standardized platelet lysates to the brain may offer an affordable treatment for CNS disorders. The potential of platelet EVs as a standalone biotherapy is another area where preclinical studies are urgently required. Both therapies could be made readily available worldwide using the domestic blood supply.

The authors would like to thank Ms. Rowan Tweedale and Dr. Odette Leiter for helpful comments on the manuscript and Mr. David Brici for assistance with the preparation of the figures and graphical abstract.

Funding support for this article was provided by the Ministry of Science and Technology (MOST) (110-2314-B-038-079), Brazil Family Foundation for Neurology, and Donald and Joan Wilson Foundation.

Contribution: T.B. and T.L.W. wrote the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Thierry Burnouf, Graduate Institute of Biomedical Materials and Tissue Engineering College of Biomedical Engineering Taipei Medical University 250 Wuxing St Xinyi District, Taipei City 110 11031, Taiwan; e-mail: thierry@tmu.edu.tw; and Tara Walker, Queensland Brain Institute, Building 79, Upland Rd, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia; e-mail: t.walker1@uq.edu.au.

1.
Gaertner
F
,
Massberg
S
.
Patrolling the vascular borders: platelets in immunity to infection and cancer
.
Nat Rev Immunol.
2019
;
19
(
12
):
747
-
760
2.
Kapur
R
,
Zufferey
A
,
Boilard
E
,
Semple
JW
.
Nouvelle cuisine: platelets served with inflammation
.
J Immunol.
2015
;
194
(
12
):
5579
-
5587
.
3.
Maouia
A
,
Rebetz
J
,
Kapur
R
,
Semple
JW
.
The immune nature of platelets revisited
.
Transfus Med Rev.
2020
;
34
(
4
):
209
-
220
.
4.
Marcoux
G
,
Laroche
A
,
Espinoza Romero
J
,
Boilard
E
.
Role of platelets and megakaryocytes in adaptive immunity
.
Platelets.
2021
;
32
(
3
):
340
-
351
.
5.
Aslam
R
,
Speck
ER
,
Kim
M
, et al
.
Platelet Toll-like receptor expression modulates lipopolysaccharide-induced thrombocytopenia and tumor necrosis factor-alpha production in vivo
.
Blood.
2006
;
107
(
2
):
637
-
641
.
6.
Henn
V
,
Slupsky
JR
,
Gräfe
M
, et al
.
CD40 ligand on activated platelets triggers an inflammatory reaction of endothelial cells
.
Nature.
1998
;
391
(
6667
):
591
-
594
.
7.
Verschoor
A
,
Neuenhahn
M
,
Navarini
AA
, et al
.
A platelet-mediated system for shuttling blood-borne bacteria to CD8α+ dendritic cells depends on glycoprotein GPIb and complement C3
.
Nat Immunol.
2011
;
12
(
12
):
1194
-
1201
.
8.
Chapman
LM
,
Aggrey
AA
,
Field
DJ
, et al
.
Platelets present antigen in the context of MHC class I
.
J Immunol.
2012
;
189
(
2
):
916
-
923
.
9.
Iannacone
M
,
Sitia
G
,
Isogawa
M
, et al
.
Platelets mediate cytotoxic T lymphocyte-induced liver damage
.
Nat Med.
2005
;
11
(
11
):
1167
-
1169
.
10.
Zufferey
A
,
Schvartz
D
,
Nolli
S
,
Reny
JL
,
Sanchez
JC
,
Fontana
P
.
Characterization of the platelet granule proteome: evidence of the presence of MHC1 in alpha-granules
.
J Proteomics.
2014
;
101
:
130
-
140
.
11.
Huang
J
,
Swieringa
F
,
Solari
FA
, et al
.
Assessment of a complete and classified platelet proteome from genome-wide transcripts of human platelets and megakaryocytes covering platelet functions
.
Sci Rep.
2021
;
11
(
1
):
12358
.
12.
Coppinger
JA
,
Maguire
PB
.
Insights into the platelet releasate
.
Curr Pharm Des.
2007
;
13
(
26
):
2640
-
2646
.
13.
Italiano
JE
Jr
,
Richardson
JL
,
Patel-Hett
S
, et al
.
Angiogenesis is regulated by a novel mechanism: pro- and antiangiogenic proteins are organized into separate platelet α granules and differentially released
.
Blood.
2008
;
111
(
3
):
1227
-
1233
.
14.
Heijnen
HF
,
Schiel
AE
,
Fijnheer
R
,
Geuze
HJ
,
Sixma
JJ
.
Activated platelets release two types of membrane vesicles: microvesicles by surface shedding and exosomes derived from exocytosis of multivesicular bodies and alpha-granules
.
Blood.
1999
;
94
(
11
):
3791
-
3799
.
15.
Wolf
P
.
The nature and significance of platelet products in human plasma
.
Br J Haematol.
1967
;
13
(
3
):
269
-
288
.
16.
Brisson
AR
,
Tan
S
,
Linares
R
,
Gounou
C
,
Arraud
N
.
Extracellular vesicles from activated platelets: a semiquantitative cryo-electron microscopy and immuno-gold labeling study
.
Platelets.
2017
;
28
(
3
):
263
-
271
.
17.
Puhm
F
,
Boilard
E
,
Machlus
KR
.
Platelet extracellular vesicles: beyond the blood
.
Arterioscler Thromb Vasc Biol.
2021
;
41
(
1
):
87
-
96
.
18.
Cauwenberghs
S
,
Feijge
MA
,
Harper
AG
,
Sage
SO
,
Curvers
J
,
Heemskerk
JW
.
Shedding of procoagulant microparticles from unstimulated platelets by integrin-mediated destabilization of actin cytoskeleton
.
FEBS Lett.
2006
;
580
(
22
):
5313
-
5320
.
19.
Aatonen
MT
,
Ohman
T
,
Nyman
TA
,
Laitinen
S
,
Grönholm
M
,
Siljander
PR
.
Isolation and characterization of platelet-derived extracellular vesicles
.
J Extracell Vesicles.
2014
;
3
(
1
)
24692
.
20.
Sandberg
H
,
Bode
AP
,
Dombrose
FA
,
Hoechli
M
,
Lentz
BR
.
Expression of coagulant activity in human platelets: release of membranous vesicles providing platelet factor 1 and platelet factor 3
.
Thromb Res.
1985
;
39
(
1
):
63
-
79
.
21.
Milioli
M
,
Ibáñez-Vea
M
,
Sidoli
S
,
Palmisano
G
,
Careri
M
,
Larsen
MR
.
Quantitative proteomics analysis of platelet-derived microparticles reveals distinct protein signatures when stimulated by different physiological agonists
.
J Proteomics.
2015
;
121
:
56
-
66
.
22.
Shai
E
,
Rosa
I
,
Parguiña
AF
,
Motahedeh
S
,
Varon
D
,
García
Á
.
Comparative analysis of platelet-derived microparticles reveals differences in their amount and proteome depending on the platelet stimulus
.
J Proteomics.
2012
;
76
(
Spec No
):
287
-
296
.
23.
Ribeiro
MF
,
Zhu
H
,
Millard
RW
,
Fan
GC
.
Exosomes function in pro- and anti-angiogenesis
.
Curr Angiogenes.
2013
;
2
(
1
):
54
-
59
.
24.
Boudreau
LH
,
Duchez
AC
,
Cloutier
N
, et al
.
Platelets release mitochondria serving as substrate for bactericidal group IIA-secreted phospholipase A2 to promote inflammation
[published correction appears in Blood. 2015;125(5):890].
Blood.
2014
;
124
(
14
):
2173
-
2183
.
25.
Leiter
O
,
Walker
TL
.
Platelets: the missing link between the blood and brain?
Prog Neurobiol.
2019
;
183
:
101695
.
26.
Goubau
C
,
Buyse
GM
,
Di Michele
M
,
Van Geet
C
,
Freson
K
.
Regulated granule trafficking in platelets and neurons: a common molecular machinery
.
Eur J Paediatr Neurol.
2013
;
17
(
2
):
117
-
125
.
27.
Kopeikina
E
,
Ponomarev
ED
.
The role of platelets in the stimulation of neuronal synaptic plasticity, electric activity, and oxidative phosphorylation: possibilities for new therapy of neurodegenerative diseases
.
Front Cell Neurosci.
2021
;
15
:
680126
.
28.
Ponomarev
ED
.
Fresh evidence for platelets as neuronal and innate immune cells: their role in the activation, differentiation, and deactivation of Th1, Th17, and tregs during tissue inflammation
.
Front Immunol.
2018
;
9
:
406
.
29.
Reed
GL
,
Fitzgerald
ML
,
Polgár
J
.
Molecular mechanisms of platelet exocytosis: insights into the “secrete” life of thrombocytes
.
Blood.
2000
;
96
(
10
):
3334
-
3342
.
30.
Wüthrich
C
,
Deranleau
DA
,
Dubler
D
,
Lüscher
EF
.
Human blood platelet secretion: optical multichannel analyzer measurements using acriflavine as a release indicator
.
Biochemistry.
1984
;
23
(
6
):
1224
-
1229
.
31.
Maclean
JA
,
Schoenwaelder
SM
. Serotonin in platelets. In:
Pilowsky
PM
, ed.
Serotonin.
Cambridge, MA
:
Academic Press
;
2019
:
91
-
119.
32.
Begni
B
,
Tremolizzo
L
,
D’Orlando
C
, et al
.
Substrate-induced modulation of glutamate uptake in human platelets
.
Br J Pharmacol.
2005
;
145
(
6
):
792
-
799
.
33.
Kaneez
FS
,
Saeed
SA
.
Investigating GABA and its function in platelets as compared to neurons
.
Platelets.
2009
;
20
(
5
):
328
-
333
.
34.
Tseng
WL
,
Chen
TH
,
Huang
CC
, et al
.
Impaired thrombin generation in reelin-deficient mice: a potential role of plasma reelin in hemostasis
.
J Thromb Haemost.
2014
;
12
(
12
):
2054
-
2064
.
35.
Gowert
NS
,
Krüger
I
,
Klier
M
, et al
.
Loss of reelin protects mice against arterial thrombosis by impairing integrin activation and thrombus formation under high shear conditions
.
Cell Signal.
2017
;
40
:
210
-
221
.
36.
Yamamoto
H
,
Gurney
ME
.
Human platelets contain brain-derived neurotrophic factor
.
J Neurosci.
1990
;
10
(
11
):
3469
-
3478
.
37.
Chacón-Fernández
P
,
Säuberli
K
,
Colzani
M
,
Moreau
T
,
Ghevaert
C
,
Barde
YA
.
Brain-derived neurotrophic factor in megakaryocytes
.
J Biol Chem.
2016
;
291
(
19
):
9872
-
9881
.
38.
Amadio
P
,
Porro
B
,
Sandrini
L
, et al
.
Patho-physiological role of BDNF in fibrin clotting
.
Sci Rep.
2019
;
9
(
1
):
389
.
39
Radka
SF
,
Holst
PA
,
Fritsche
M
,
Altar
CA
.
Presence of brain-derived neurotrophic factor in brain and human and rat but not mouse serum detected by a sensitive and specific immunoassay
.
Brain Res.
1996
;
709
(
1
):
122
-
301
.
40.
Nebie
O
,
Carvalho
K
,
Barro
L
, et al
.
Human platelet lysate biotherapy for traumatic brain injury: preclinical assessment
.
Brain.
2021
;
144
(
10
):
3142
-
3158
.
41.
Nurden
AT
.
The biology of the platelet with special reference to inflammation, wound healing and immunity
.
Front Biosci.
2018
;
23
(
4
):
726
-
751
.
42.
Blair
P
,
Flaumenhaft
R
.
Platelet alpha-granules: basic biology and clinical correlates
.
Blood Rev.
2009
;
23
(
4
):
177
-
189
.
43.
Filippov
V
,
Kronenberg
G
,
Pivneva
T
, et al
.
Subpopulation of nestin-expressing progenitor cells in the adult murine hippocampus shows electrophysiological and morphological characteristics of astrocytes
.
Mol Cell Neurosci.
2003
;
23
(
3
):
373
-
382
.
44.
Mirzadeh
Z
,
Merkle
FT
,
Soriano-Navarro
M
,
Garcia-Verdugo
JM
,
Alvarez-Buylla
A
.
Neural stem cells confer unique pinwheel architecture to the ventricular surface in neurogenic regions of the adult brain
.
Cell Stem Cell.
2008
;
3
(
3
):
265
-
278
.
45.
Moss
J
,
Gebara
E
,
Bushong
EA
, et al
.
Fine processes of nestin-GFP-positive radial glia-like stem cells in the adult dentate gyrus ensheathe local synapses and vasculature
.
Proc Natl Acad Sci USA.
2016
;
113
(
18
):
E2536
-
E2545
.
46.
Kazanis
I
,
Feichtner
M
,
Lange
S
, et al
.
Lesion-induced accumulation of platelets promotes survival of adult neural stem/progenitor cells
.
Exp Neurol.
2015
;
269
:
75
-
89
.
47.
Hayon
Y
,
Dashevsky
O
,
Shai
E
,
Varon
D
,
Leker
RR
.
Platelet microparticles promote neural stem cell proliferation, survival, and differentiation
.
J Mol Neurosci.
2012
;
47
(
3
):
659
-
665
.
48.
Hayon
Y
,
Dashevsky
O
,
Shai
E
,
Brill
A
,
Varon
D
,
Leker
RR
.
Platelet microparticles induce angiogenesis and neurogenesis after cerebral ischemia
.
Curr Neurovasc Res.
2012
;
9
(
3
):
185
-
192
.
49.
Hayon
Y
,
Dashevsky
O
,
Shai
E
,
Varon
D
,
Leker
RR
.
Platelet lysates stimulate angiogenesis, neurogenesis, and neuroprotection after stroke
.
Thromb Haemost.
2013
;
110
(
2
):
323
-
330
.
50.
Leiter
O
,
Seidemann
S
,
Overall
RW
, et al
.
Exercise-induced activated platelets increase adult hippocampal precursor proliferation and promote neuronal differentiation
.
Stem Cell Reports.
2019
;
12
(
4
):
667
-
679
.
51.
Niiya
K
,
Hodson
E
,
Bader
R
, et al
.
Increased surface expression of the membrane glycoprotein IIb/IIIa complex induced by platelet activation. Relationship to the binding of fibrinogen and platelet aggregation
.
Blood.
1987
;
70
(
2
):
475
-
483
.
52.
Dukhinova
M
,
Kuznetsova
I
,
Kopeikina
E
, et al
.
Platelets mediate protective neuroinflammation and promote neuronal plasticity at the site of neuronal injury
.
Brain Behav Immun.
2018
;
74
:
7
-
27
.
53.
Sotnikov
I
,
Veremeyko
T
,
Starossom
SC
,
Barteneva
N
,
Weiner
HL
,
Ponomarev
ED
.
Platelets recognize brain-specific glycolipid structures, respond to neurovascular damage and promote neuroinflammation
[published correction appears in PLoS One. 2014;9(1):10].
PLoS One.
2013
;
8
(
3
):
e58979
.
54.
Kopeikina
E
,
Dukhinova
M
,
Yung
AWY
, et al
.
Platelets promote epileptic seizures by modulating brain serotonin level, enhancing neuronal electric activity, and contributing to neuroinflammation and oxidative stress
.
Prog Neurobiol.
2020
;
188
:
101783
.
55.
Wieraszko
A
,
Li
G
,
Kornecki
E
,
Hogan
MV
,
Ehrlich
YH
.
Long-term potentiation in the hippocampus induced by platelet-activating factor
.
Neuron.
1993
;
10
(
3
):
553
-
557
.
56.
Ransohoff
RM
.
How neuroinflammation contributes to neurodegeneration
.
Science.
2016
;
353
(
6301
):
777
-
783
.
57.
Guzman-Martinez
L
,
Maccioni
RB
,
Andrade
V
,
Navarrete
LP
,
Pastor
MG
,
Ramos-Escobar
N
.
Neuroinflammation as a common feature of neurodegenerative disorders
.
Front Pharmacol.
2019
;
10
:
1008
.
58.
Page
MJ
,
Pretorius
E
.
Platelet behavior contributes to neuropathologies: a focus on Alzheimer’s and Parkinson’s disease
.
Semin Thromb Hemost.
2021
;
48
(
3
):
382
-
404
.
59.
Orian
JM
,
D’Souza
CS
,
Kocovski
P
, et al
.
Platelets in multiple sclerosis: early and central mediators of inflammation and neurodegeneration and attractive targets for molecular imaging and site-directed therapy
.
Front Immunol.
2021
;
12
:
620963
.
60.
Segura-Collar
B
,
Mata-Martínez
P
,
Hernández-Laín
A
,
Sánchez-Gómez
P
,
Gargini
R
.
Blood-brain barrier disruption: a common driver of central nervous system diseases
.
Neuroscientist.
2022
;
28
(
3
):
222
-
237
.
61.
Leiter
O
,
Walker
TL
.
Platelets in neurodegenerative conditions-friend or foe?
Front Immunol.
2020
;
11
:
747
.
62.
Thomas
MR
,
Storey
RF
.
The role of platelets in inflammation
.
Thromb Haemost.
2015
;
114
(
3
):
449
-
458
.
63.
Margraf
A
,
Zarbock
A
.
Platelets in inflammation and resolution
.
J Immunol.
2019
;
203
(
9
):
2357
-
2367
.
64.
Sang
Y
,
Roest
M
,
de Laat
B
,
de Groot
PG
,
Huskens
D
.
Interplay between platelets and coagulation
.
Blood Rev.
2021
;
46
:
100733
.
65.
French
SL
,
Butov
KR
,
Allaeys
I
, et al
.
Platelet-derived extracellular vesicles infiltrate and modify the bone marrow during inflammation
.
Blood Adv.
2020
;
4
(
13
):
3011
-
3023
.
66.
Kuravi
SJ
,
Harrison
P
,
Rainger
GE
,
Nash
GB
.
Ability of platelet-derived extracellular vesicles to promote neutrophil-endothelial cell interactions
.
Inflammation.
2019
;
42
(
1
):
290
-
305
.
67.
Horstman
LL
,
Jy
W
,
Ahn
YS
, et al
.
Role of platelets in neuroinflammation: a wide-angle perspective
.
J Neuroinflammation.
2010
;
7
(
1
):
10
.
68.
Cloutier
N
,
Allaeys
I
,
Marcoux
G
, et al
.
Platelets release pathogenic serotonin and return to circulation after immune complex-mediated sequestration
.
Proc Natl Acad Sci USA.
2018
;
115
(
7
):
E1550
-
E1559
.
69.
Jones
LD
,
Jackson
JW
,
Maggirwar
SB
.
Modeling HIV-1 induced neuroinflammation in mice: role of platelets in mediating blood-brain barrier dysfunction
.
PLoS One.
2016
;
11
(
3
):
e0151702
.
70.
Schleicher
RI
,
Reichenbach
F
,
Kraft
P
, et al
.
Platelets induce apoptosis via membrane-bound FasL
[published correction appears in Blood. 2016;127(2):270].
Blood.
2015
;
126
(
12
):
1483
-
1493
.
71.
Kocovski
P
,
Jiang
X
,
D’Souza
CS
, et al
.
Platelet depletion is effective in ameliorating anxiety-like behavior and reducing the pro-inflammatory environment in the hippocampus in murine experimental autoimmune encephalomyelitis
.
J Clin Med.
2019
;
8
(
2
):
162
.
72.
Kniewallner
KM
,
de Sousa
DMB
,
Unger
MS
,
Mrowetz
H
,
Aigner
L
.
Platelets in amyloidogenic mice are activated and invade the brain
.
Front Neurosci.
2020
;
14
:
129
.
73.
Bhat
SA
,
Goel
R
,
Shukla
R
,
Hanif
K
.
Platelet CD40L induces activation of astrocytes and microglia in hypertension
.
Brain Behav Immun.
2017
;
59
:
173
-
189
.
74.
Giles
JA
,
Greenhalgh
AD
,
Denes
A
, et al
.
Neutrophil infiltration to the brain is platelet-dependent, and is reversed by blockade of platelet GPIbα
.
Immunology.
2018
;
154
(
2
):
322
-
328
.
75.
Rawish
E
,
Nording
H
,
Münte
T
,
Langer
HF
.
Platelets as mediators of neuroinflammation and thrombosis
.
Front Immunol.
2020
;
11
:
548631
.
76.
Vogelsang
A
,
Eichler
S
,
Huntemann
N
, et al
.
Platelet inhibition by low-dose acetylsalicylic acid reduces neuroinflammation in an animal model of multiple sclerosis
.
Int J Mol Sci.
2021
;
22
(
18
):
9915
.
77.
Canobbio
I
.
Blood platelets: circulating mirrors of neurons?
Res Pract Thromb Haemost.
2019
;
3
(
4
):
564
-
565
.
78.
Behari
M
,
Shrivastava
M
.
Role of platelets in neurodegenerative diseases: a universal pathophysiology
.
Int J Neurosci.
2013
;
123
(
5
):
287
-
299
.
79.
Espinosa-Parrilla
Y
,
Gonzalez-Billault
C
,
Fuentes
E
,
Palomo
I
,
Alarcón
M
.
Decoding the role of platelets and related microRNAs in aging and neurodegenerative disorders
.
Front Aging Neurosci.
2019
;
11
:
151
.
80.
Ferrer-Raventós
P
,
Beyer
K
.
Alternative platelet activation pathways and their role in neurodegenerative diseases
.
Neurobiol Dis.
2021
;
159
:
105512
.
81.
Li
Q-X
,
Evin
G
,
Small
DH
,
Multhaup
G
,
Beyreuther
K
,
Masters
CL
.
Proteolytic processing of Alzheimer’s disease β A4 amyloid precursor protein in human platelets
.
J Biol Chem.
1995
;
270
(
23
):
14140
-
14147
.
82.
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
.
83.
Veitinger
M
,
Varga
B
,
Guterres
SB
,
Zellner
M
.
Platelets, a reliable source for peripheral Alzheimer’s disease biomarkers?
Acta Neuropathol Commun.
2014
;
2
(
1
):
65
.
84.
Inyushin
M
,
Zayas-Santiago
A
,
Rojas
L
,
Kucheryavykh
L
.
On the role of platelet-generated amyloid beta peptides in certain amyloidosis health complications
.
Front Immunol.
2020
;
11
:
571083
.
85.
Evin
G
,
Li
Q-X
.
Platelets and Alzheimer’s disease: potential of APP as a biomarker
.
World J Psychiatry.
2012
;
2
(
6
):
102
-
113
.
86.
Bush
AI
,
Martins
RN
,
Rumble
B
, et al
.
The amyloid precursor protein of Alzheimer’s disease is released by human platelets
.
J Biol Chem.
1990
;
265
(
26
):
15977
-
15983
.
87.
Smirnov
A
,
Trupp
A
,
Henkel
AW
, et al
.
Differential processing and secretion of Abeta peptides and sAPPalpha in human platelets is regulated by thrombin and prostaglandine 2
.
Neurobiol Aging.
2009
;
30
(
10
):
1552
-
1562
.
88.
Davies
TA
,
Long
HJ
,
Sgro
K
, et al
.
Activated Alzheimer disease platelets retain more beta amyloid precursor protein
.
Neurobiol Aging.
1997
;
18
(
2
):
147
-
153
.
89.
Soscia
SJ
,
Kirby
JE
,
Washicosky
KJ
, et al
.
The Alzheimer’s disease-associated amyloid beta-protein is an antimicrobial peptide
.
PLoS One.
2010
;
5
(
3
):
e9505
.
90.
Kumar
DKV
,
Choi
SH
,
Washicosky
KJ
, et al
.
Amyloid-β peptide protects against microbial infection in mouse and worm models of Alzheimer’s disease
.
Science Transl Med.
2016
;
8
(
340
):
340ra372
.
91.
Zlokovic
BV
,
Martel
CL
,
Mackic
JB
, et al
.
Brain uptake of circulating apolipoproteins J and E complexed to Alzheimer’s amyloid beta
.
Biochem Biophys Res Commun.
1994
;
205
(
2
):
1431
-
1437
.
92.
Donner
L
,
Fälker
K
,
Gremer
L
, et al
.
Platelets contribute to amyloid-β aggregation in cerebral vessels through integrin αIIbβ3-induced outside-in signaling and clusterin release
.
Sci Signal.
2016
;
9
(
429
):
ra52
.
93.
Shen
MY
,
Hsiao
G
,
Fong
TH
, et al
.
Amyloid beta peptide-activated signal pathways in human platelets
.
Eur J Pharmacol.
2008
;
588
(
2-3
):
259
-
266
.
94.
Wu
T
,
Chen
L
,
Zhou
L
,
Xu
J
,
Guo
K
.
Platelets transport β-amyloid from the peripheral blood into the brain by destroying the blood-brain barrier to accelerate the process of Alzheimer’s disease in mouse models
.
Aging (Albany NY).
2021
;
13
(
5
):
7644
-
7659
.
95.
Canobbio
I
,
Visconte
C
,
Oliviero
B
, et al
.
Increased platelet adhesion and thrombus formation in a mouse model of Alzheimer’s disease
.
Cell Signal.
2016
;
28
(
12
):
1863
-
1871
.
96.
Jarre
A
,
Gowert
NS
,
Donner
L
, et al
.
Pre-activated blood platelets and a pro-thrombotic phenotype in APP23 mice modeling Alzheimer’s disease
.
Cell Signal.
2014
;
26
(
9
):
2040
-
2050
.
97.
Schmaier
AH
.
Alzheimer disease is in part a thrombohemorrhagic disorder
.
J Thromb Haemost.
2016
;
14
(
5
):
991
-
994
.
98.
Bu
XL
,
Xiang
Y
,
Jin
WS
, et al
.
Blood-derived amyloid-β protein induces Alzheimer’s disease pathologies
.
Mol Psychiatry.
2018
;
23
(
9
):
1948
-
1956
.
99.
Zhao
Y
,
Qian
R
,
Zhang
J
, et al
.
Young blood plasma reduces Alzheimer’s disease-like brain pathologies and ameliorates cognitive impairment in 3×Tg-AD mice
.
Alzheimers Res Ther.
2020
;
12
(
1
):
70
.
100.
Middeldorp
J
,
Lehallier
B
,
Villeda
SA
, et al
.
Preclinical assessment of young blood plasma for Alzheimer disease
.
JAMA Neurol.
2016
;
73
(
11
):
1325
-
1333
.
101.
Sha
SJ
,
Deutsch
GK
,
Tian
L
, et al
.
Safety, tolerability, and feasibility of young plasma infusion in the plasma for Alzheimer symptom amelioration study: a randomized clinical trial
.
JAMA Neurol.
2019
;
76
(
1
):
35
-
40
.
102.
Boada
M
,
López
OL
,
Olazarán
J
, et al
.
A randomized, controlled clinical trial of plasma exchange with albumin replacement for Alzheimer’s disease: primary results of the AMBAR Study
.
Alzheimers Dement.
2020
;
16
(
10
):
1412
-
1425
.
103.
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 (Vienna).
2012
;
119
(
7
):
815
-
819
.
104.
Farías
G
,
Pérez
P
,
Slachevsky
A
,
Maccioni
RB
.
Platelet tau pattern correlates with cognitive status in Alzheimer’s disease
.
J Alzheimers Dis.
2012
;
31
(
1
):
65
-
69
.
105.
Neumann
K
,
Farías
G
,
Slachevsky
A
,
Perez
P
,
Maccioni
RB
.
Human platelets tau: a potential peripheral marker for Alzheimer’s disease
.
J Alzheimers Dis.
2011
;
25
(
1
):
103
-
109
.
106.
Shi
Y
,
Gu
L
,
Wang
Q
, et al
.
Platelet amyloid-β protein precursor (AβPP) ratio and phosphorylated tau as promising indicators for early Alzheimer’s disease
.
J Gerontol A Biol Sci Med Sci.
2020
;
75
(
4
):
664
-
670
.
107.
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
.
108.
Bentham
P
,
Gray
R
,
Sellwood
E
,
Hills
R
,
Crome
P
,
Raftery
J
;
AD2000 Collaborative Group
.
Aspirin in Alzheimer’s disease (AD2000): a randomised open-label trial
.
Lancet Neurol.
2008
;
7
(
1
):
41
-
49
.
109.
Ryan
J
,
Storey
E
,
Murray
AM
, et al;
ASPREE Investigator Group
.
Randomized placebo-controlled trial of the effects of aspirin on dementia and cognitive decline
.
Neurology.
2020
;
95
(
3
):
e320
-
e331
.
110.
Sakurai
H
,
Hanyu
H
,
Sato
T
, et al
.
Effects of cilostazol on cognition and regional cerebral blood flow in patients with Alzheimer’s disease and cerebrovascular disease: a pilot study
.
Geriatr Gerontol Int.
2013
;
13
(
1
):
90
-
97
.
111.
Tai
S-Y
,
Chen
C-H
,
Chien
C-Y
,
Yang
Y-H
.
Cilostazol as an add-on therapy for patients with Alzheimer’s disease in Taiwan: a case control study
.
BMC Neurol.
2017
;
17
(
1
):
40
.
112.
Saito
S
,
Kojima
S
,
Oishi
N
, et al
.
A multicenter, randomized, placebo-controlled trial for cilostazol in patients with mild cognitive impairment: the COMCID study protocol
.
Alzheimers Dement (N Y).
2016
;
2
(
4
):
250
-
257
.
113.
Raadsen
M
,
Du Toit
J
,
Langerak
T
,
van Bussel
B
,
van Gorp
E
,
Goeijenbier
M
.
Thrombocytopenia in virus infections
.
J Clin Med.
2021
;
10
(
4
):
877
.
114.
Wassmer
SC
,
Combes
V
,
Candal
FJ
,
Juhan-Vague
I
,
Grau
GE
.
Platelets potentiate brain endothelial alterations induced by plasmodium falciparum
.
Infect Immun.
2006
;
74
(
1
):
645
-
653
.
115.
Randriamboavonjy
V
,
Sopova
K
,
Stellos
K
,
Laske
C
.
Platelets as potential link between diabetes and Alzheimer’s disease
.
Curr Alzheimer Res.
2014
;
11
(
9
):
862
-
868
.
116.
Schallmoser
K
,
Henschler
R
,
Gabriel
C
,
Koh
MBC
,
Burnouf
T
.
Production and quality requirements of human platelet lysate: a position statement from the working party on cellular therapies of the International Society of Blood Transfusion
.
Trends Biotechnol.
2020
;
38
(
1
):
13
-
23
.
117.
Nebie
O
,
Devos
D
,
Vingtdeux
V
, et al
.
The neuroprotective activity of heat-treated human platelet lysate biomaterials manufactured from outdated pathogen-reduced (amotosalen/UVA) platelet concentrates
.
J Biomed Sci.
2019
;
26
(
1
):
89
.
118.
Burnouf
T
.
Modern plasma fractionation
.
Transfus Med Rev.
2007
;
21
(
2
):
101
-
117
.
119.
Chou
ML
,
Wu
JW
,
Gouel
F
, et al
.
Tailor-made purified human platelet lysate concentrated in neurotrophins for treatment of Parkinson’s disease
.
Biomaterials.
2017
;
142
:
77
-
89
.
120.
Anitua
E
,
Pascual
C
,
Antequera
D
, et al
.
Plasma rich in growth factors (PRGF-Endoret) reduces neuropathologic hallmarks and improves cognitive functions in an Alzheimer’s disease mouse model
.
Neurobiol Aging.
2014
;
35
(
7
):
1582
-
1595
.
121.
Anitua
E
,
Pascual
C
,
Pérez-Gonzalez
R
, et al
.
Intranasal delivery of plasma and platelet growth factors using PRGF-Endoret system enhances neurogenesis in a mouse model of Alzheimer’s disease
.
PLoS One.
2013
;
8
(
9
):
e73118
.
122.
Anitua
E
,
Pascual
C
,
Pérez-Gonzalez
R
,
Orive
G
,
Carro
E
.
Intranasal PRGF-Endoret enhances neuronal survival and attenuates NF-κB-dependent inflammation process in a mouse model of Parkinson’s disease
.
J Control Release.
2015
;
203
:
170
-
180
.
123.
Gouel
F
,
Do Van
B
,
Chou
ML
, et al
.
The protective effect of human platelet lysate in models of neurodegenerative disease: involvement of the Akt and MEK pathways
.
J Tissue Eng Regen Med.
2017
;
11
(
11
):
3236
-
3240
.
124.
Gouel
F
,
Rolland
AS
,
Devedjian
JC
,
Burnouf
T
,
Devos
D
.
Past and future of neurotrophic growth factors therapies in ALS: from single neurotrophic growth factor to stem cells and human platelet lysates
.
Front Neurol.
2019
;
10
:
835
.
125.
Nebie
O
,
Barro
L
,
Wu
YW
, et al
.
Heat-treated human platelet pellet lysate modulates microglia activation, favors wound healing and promotes neuronal differentiation in vitro.
Platelets.
2021
;
32
(
2
):
226
-
237
.
126.
Gouel
F
,
Timmerman
K
,
Gosset
P
, et al
.
Whole and fractionated human platelet lysate biomaterials-based biotherapy induces strong neuroprotection in experimental models of amyotrophic lateral sclerosis
.
Biomaterials.
2022
;
280
:
121311
.
127.
Agrahari
V
,
Burnouf
PA
,
Burnouf
T
,
Agrahari
V
.
Nanoformulation properties, characterization, and behavior in complex biological matrices: challenges and opportunities for brain-targeted drug delivery applications and enhanced translational potential
.
Adv Drug Deliv Rev.
2019
;
148
:
146
-
180
.
128.
Cone
AS
,
Yuan
X
,
Sun
L
, et al
.
Mesenchymal stem cell-derived extracellular vesicles ameliorate Alzheimer’s disease-like phenotypes in a preclinical mouse model
.
Theranostics.
2021
;
11
(
17
):
8129
-
8142
.
129.
Johnson
J
,
Wu
YW
,
Blyth
C
,
Lichtfuss
G
,
Goubran
H
,
Burnouf
T
.
Prospective therapeutic applications of platelet extracellular vesicles
.
Trends Biotechnol.
2021
;
39
(
6
):
598
-
612
.
130.
Nyam-Erdene
A
,
Nebie
O
,
Delila
L
, et al
.
Characterization and chromatographic isolation of platelet extracellular vesicles from human platelet lysates for applications in neuroregenerative medicine
.
ACS Biomater Sci Eng.
2021
;
7
(
12
):
5823
-
5835
.
131.
Melki
I
,
Tessandier
N
,
Zufferey
A
,
Boilard
E
.
Platelet microvesicles in health and disease
.
Platelets.
2017
;
28
(
3
):
214
-
221
.
132.
Ferreira
PM
,
Bozbas
E
,
Tannetta
SD
, et al
.
Mode of induction of platelet-derived extracellular vesicles is a critical determinant of their phenotype and function
.
Sci Rep.
2020
;
10
(
1
):
18061
.
133.
Narbute
K
,
Piļipenko
V
,
Pupure
J
, et al
.
Intranasal administration of extracellular vesicles derived from human teeth stem cells improves motor symptoms and normalizes tyrosine hydroxylase expression in the substantia nigra and striatum of the 6-hydroxydopamine-treated rats
.
Stem Cells Transl Med.
2019
;
8
(
5
):
490
-
499
.
134.
Ciabattoni
G
,
Porreca
E
,
Di Febbo
C
, et al
.
Determinants of platelet activation in Alzheimer’s disease
.
Neurobiol Aging.
2007
;
28
(
3
):
336
-
342
.
135.
Wiest
I
,
Wiemers
T
,
Kraus
MJ
, et al
.
Multivariate platelet analysis differentiates between patients with Alzheimer’s disease and healthy controls at first clinical diagnosis
.
J Alzheimers Dis.
2019
;
71
(
3
):
993
-
1004
.
136.
Kniewallner
KM
,
Wenzel
D
,
Humpel
C
.
Thiazine red(+) platelet inclusions in cerebral blood vessels are first signs in an Alzheimer’s disease mouse model
.
Sci Rep.
2016
;
6
(
1
):
28447
.
137.
Fišar
Z
,
Jirák
R
,
Zvěřová
M
, et al
.
Plasma amyloid beta levels and platelet mitochondrial respiration in patients with Alzheimer’s disease
.
Clin Biochem.
2019
;
72
:
71
-
80
.
138.
Parker
WD
Jr
,
Filley
CM
,
Parks
JK
.
Cytochrome oxidase deficiency in Alzheimer’s disease
.
Neurology.
1990
;
40
(
8
):
1302
-
1303
.
139.
Swerdlow
RH
,
Koppel
S
,
Weidling
I
,
Hayley
C
,
Ji
Y
,
Wilkins
HM
.
Mitochondria, cybrids, aging, and Alzheimer’s disease
.
Prog Mol Biol Transl Sci.
2017
;
146
:
259
-
302
.
140.
Johnston
JA
,
Liu
WW
,
Coulson
DT
, et al
.
Platelet beta-secretase activity is increased in Alzheimer’s disease
.
Neurobiol Aging.
2008
;
29
(
5
):
661
-
668
.
141.
Norman
TR
,
Chiu
E
,
French
MA
.
Platelet monoamine oxidase activity in patients with Huntington’s disease
.
Clin Exp Pharmacol Physiol.
1987
;
14
(
6
):
547
-
550
.
142.
Forlenza
OV
,
Torres
CA
,
Talib
LL
, et al
.
Increased platelet GSK3B activity in patients with mild cognitive impairment and Alzheimer’s disease
.
J Psychiatr Res.
2011
;
45
(
2
):
220
-
224
.
143.
Ferrarese
C
,
Begni
B
,
Canevari
C
, et al
.
Glutamate uptake is decreased in platelets from Alzheimer’s disease patients
.
Ann Neurol.
2000
;
47
(
5
):
641
-
643
.
144.
Koçer
A
,
Yaman
A
,
Niftaliyev
E
,
Dürüyen
H
,
Eryılmaz
M
,
Koçer
E
.
Assessment of platelet indices in patients with neurodegenerative diseases: mean platelet volume was increased in patients with Parkinson’s disease
.
Curr Gerontol Geriatr Res.
2013
;
2013
:
986254
.
145.
Ferrarese
C
,
Zoia
C
,
Pecora
N
, et al
.
Reduced platelet glutamate uptake in Parkinson’s disease
.
J Neural Transm (Vienna).
1999
;
106
(
7-8
):
685
-
692
.
146.
Sala
G
,
Brighina
L
,
Saracchi
E
, et al
.
Vesicular monoamine transporter 2 mRNA levels are reduced in platelets from patients with Parkinson’s disease
.
J Neural Transm (Vienna).
2010
;
117
(
9
):
1093
-
1098
.
147.
Benecke
R
,
Strümper
P
,
Weiss
H
.
Electron transfer complexes I and IV of platelets are abnormal in Parkinson’s disease but normal in Parkinson-plus syndromes
.
Brain.
1993
;
116
(
Pt 6
):
1451
-
1463
.
148.
Haas
RH
,
Nasirian
F
,
Nakano
K
, et al
.
Low platelet mitochondrial complex I and complex II/III activity in early untreated Parkinson’s disease
.
Ann Neurol.
1995
;
37
(
6
):
714
-
722
.
149.
Meredith
GE
,
Totterdell
S
,
Beales
M
,
Meshul
CK
.
Impaired glutamate homeostasis and programmed cell death in a chronic MPTP mouse model of Parkinson’s disease
.
Exp Neurol.
2009
;
219
(
1
):
334
-
340
.
150.
Denis
HL
,
Lamontagne-Proulx
J
,
St-Amour
I
, et al
.
Platelet abnormalities in Huntington’s disease
J Neurol Neurosurg Psychiatry.
2019
;
90
(
3
):
272
-
283
.
151.
Reilmann
R
,
Rolf
LH
,
Lange
HW
.
Huntington’s disease: N-methyl-D-aspartate receptor coagonist glycine is increased in platelets
.
Exp Neurol.
1997
;
144
(
2
):
416
-
419
.
152.
Maglione
V
,
Giallonardo
P
,
Cannella
M
,
Martino
T
,
Frati
L
,
Squitieri
F
.
Adenosine A2A receptor dysfunction correlates with age at onset anticipation in blood platelets of subjects with Huntington’s disease
.
Am J Med Genet B Neuropsychiatr Genet.
2005
;
139B
(
1
):
101
-
105
.
153.
Carrizzo
A
,
Di Pardo
A
,
Maglione
V
, et al
.
Nitric oxide dysregulation in platelets from patients with advanced Huntington disease
.
PLoS One.
2014
;
9
(
2
):
e89745
.
154.
Shrivastava
M
,
Vivekanandhan
S
,
Pati
U
,
Behari
M
,
Das
TK
.
Mitochondrial perturbance and execution of apoptosis in platelet mitochondria of patients with amyotrophic lateral sclerosis
.
Int J Neurosci.
2011
;
121
(
3
):
149
-
158
.
155.
Shrivastava
M
,
Das
TK
,
Behari
M
,
Pati
U
,
Vivekanandhan
S
.
Ultrastructural variations in platelets and platelet mitochondria: a novel feature in amyotrophic lateral sclerosis
.
Ultrastruct Pathol.
2011
;
35
(
2
):
52
-
59
.
156.
Hishizawa
M
,
Yamashita
H
,
Akizuki
M
,
Urushitani
M
,
Takahashi
R
.
TDP-43 levels are higher in platelets from patients with sporadic amyotrophic lateral sclerosis than in healthy controls
.
Neurochem Int.
2019
;
124
:
41
-
45
.
157.
Ehinger
JK
,
Morota
S
,
Hansson
MJ
,
Paul
G
,
Elmér
E
.
Mitochondrial dysfunction in blood cells from amyotrophic lateral sclerosis patients
.
J Neurol.
2015
;
262
(
6
):
1493
-
1503
.
158.
Dupuis
L
,
Spreux-Varoquaux
O
,
Bensimon
G
, et al
.
Platelet serotonin level predicts survival in amyotrophic lateral sclerosis
.
PLoS One.
2010
;
5
(
10
):
e13346
.
159.
Ferrarese
C
,
Sala
G
,
Riva
R
, et al;
Italian ALS Study Group
.
Decreased platelet glutamate uptake in patients with amyotrophic lateral sclerosis
.
Neurology.
2001
;
56
(
2
):
270
-
272
.
160.
Smirnova
IV
,
Festoff
BW
.
Alterations in serum thrombospondin in patients with amyotrophic lateral sclerosis
.
J Neurol Sci.
1994
;
127
(
2
):
207
-
213
.
161.
Sheremata
WA
,
Jy
W
,
Horstman
LL
,
Ahn
YS
,
Alexander
JS
,
Minagar
A
.
Evidence of platelet activation in multiple sclerosis
.
J Neuroinflammation.
2008
;
5
(
1
):
27
.
162.
Wright
HP
,
Thompson
RH
,
Zilkha
KJ
.
Platelet adhesiveness in multiple sclerosis
.
Lancet.
1965
;
2
(
7422
):
1109
-
1110
.
163.
Sáenz-Cuesta
M
,
Irizar
H
,
Castillo-Triviño
T
, et al
.
Circulating microparticles reflect treatment effects and clinical status in multiple sclerosis
.
Biomarkers Med.
2014
;
8
(
5
):
653
-
661
.
164.
Starossom
SC
,
Veremeyko
T
,
Yung
AW
, et al
.
Platelets play differential role during the initiation and progression of autoimmune neuroinflammation
.
Circ Res.
2015
;
117
(
9
):
779
-
792
.
165.
Gul
S
,
Smith
AD
,
Thompson
RH
,
Wright
HP
,
Zilkha
KJ
.
Fatty acid composition of phospholipids from platelets and erythrocytes in multiple sclerosis
.
J Neurol Neurosurg Psychiatry.
1970
;
33
(
4
):
506
-
510
.
Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.
Sign in via your Institution