Open access peer-reviewed chapter

Part Two: Extracellular Vesicles as a Risk Factor in Neurodegenerative Diseases

Written By

Laura Cristina Ceafalan, Octavian Costin Ioghen, Daciana Silvia Marta, Alina Constantin, Nicoleta Alexandru, Miruna Nemecz, Gabriela Tanko, Alexandru Filippi, Stefania Lucia Magda, Florina Bojin, Virgil Paunescu, Dragos Vinereanu, Adriana Georgescu and Mihaela Gherghiceanu

Submitted: 10 April 2019 Reviewed: 30 April 2019 Published: 05 July 2019

DOI: 10.5772/intechopen.86604

From the Edited Volume

Extracellular Vesicles and Their Importance in Human Health

Edited by Ana Gil De Bona and Jose Antonio Reales Calderon

Chapter metrics overview

1,252 Chapter Downloads

View Full Metrics

Abstract

Extracellular vesicles (EVs) involved in the intercellular communication hold cell-specific cargos that contain proteins, various species of RNA and lipids. EVs are emerging as powerful tools for diagnosis and therapy in most diseases but little is known about their role in central nervous system (CNS) physiology or disease. Considering the extraordinary intricated cytoarchitecture of the brain, the implication of EVs in its pathophysiology is difficult to establish. Blood circulating EVs derived from local or distant vascular cells or EVs released from brain into the cerebrospinal fluid (CSF) may influence the brain activity. EVs released in the blood stream from various tissues may influence the brain by passing through the blood-brain barrier (BBB) or through choroid plexus. Since the choroid plexus has also a clearance role, it might be possible that EVs carrying brain abnormal proteins to pass into the blood can be detected. Thus, considering that EVs are specialized cargos bearing combined signals between cells, they might be an interesting therapy target in the future for both regulating neurogenesis and abnormal protein clearance. We present here data gathered about EVs that may influence the CNS functionality and be involved in most common neurodegenerative diseases.

Keywords

  • extracellular vesicles
  • exosomes
  • intercellular communication
  • brain barriers
  • neurodegenerative diseases

1. Introduction

Extracellular vesicle (EV) is a term used to define a heterogeneous group of vesicles isolated from biological fluids or tissues. EVs seem to mediate complex cell-to-cell communication over long distances or nearby through various macromolecules: polypeptides, various species of RNAs, and/or lipids. The classification of EVs is based on their size and mechanism of biogenesis and includes: exosomes, less than 100 nm small vesicles released from multivesicular bodies after endocytosed materials have been sorted in the endolysosomal compartment [1, 2]; ectosomes, up to 500 nm larger vesicles budding from the plasma membrane [2, 3]; and multivesicular cargos, consisting of numerous vesicles, about 150 nm, enclosed in a plasma membrane-derived shield [4]. Although many medical fields experienced real progress with newly discovered diagnostic tools or treatments for several diseases, smaller steps are taken in the field of cerebrovascular and neurodegenerative diseases. Age-related changes, cardiac diseases, and atherosclerosis are known to contribute to the pathogenic mechanism of cerebrovascular and neurodegenerative diseases affecting the elderly.

Advertisement

2. Biological content of extracellular vesicles

An increasing body of evidence proves that EVs are not only involved in the waste disposal system, but, more importantly, they function as membrane-bound carriers for intercellular communication [5, 6]. This type of intercellular communication was proven to modulate cellular functions both in homeostatic and pathological conditions [6, 7]. High concentrations of EVs were detected in culture supernatants and biological fluids [8, 9]. For example, EVs have been isolated from CSF and proved to contain overrepresentation of brain-specific proteins derived from cerebral white mater and choroid plexus [10]. Several studies demonstrated that the level and composition of circulating EVs are altered in disease states, neurodegenerative diseases included [10, 11, 12]. Endothelial cells (ECs) and platelets have been most studied as sources for EVs [13, 14], but circulating cells as monocytes or lymphocytes may also be a source for EVs.

Protein composition of the EV-enclosing membrane, mainly different types of integrins, cell adhesion molecules, and tetraspanins, guides the interaction with the recipient cells, the targeting, or recruitment once EVs are released into the extracellular environment [15]. Specific molecules allow EVs either to interact with surface receptors on recipient cells to activate signaling cascades, or to promote their docking and uptake. One potential mechanism is direct membrane fusion with direct transfer of the cargo molecules into the recipient cytosol [16]. Endocytosis, including clathrin-dependent endocytosis, lipid raft-dependent pathways, phagocytosis, and even micropinocytosis, was more frequently considered [15].

A recent quantitative proteomic analysis allowing comparison of different EV subpopulations [17] proved that several classic exosome markers such as flotillin-1, heat-shock 70-kDa proteins, actin, and MHC I and II are present in all EV fractions obtained by successive centrifugation. Moreover, classic exosomal tetraspanins CD9, CD63, and CD81 were unreached in the exosomal fraction but also detected in different amounts in larger EVs. The study suggests a further classification of EV pelleting at high speed into four subcategories: (a) EVs enriched in all tetraspanins and endosome markers (bona fide exosomes); (b) EVs devoid of CD63 and CD81 but enriched in CD9 (associated with plasma membrane and an early endocytic signature); (c) EVs devoid of CD63/CD9/CD81; and (d) EVs enriched in extracellular matrix (ECM) or serum-derived factors. They also propose five categories of proteins with different relative distributions in different EV populations that relate them to their intracellular source [17]. Thus, exosomes contain ECM proteins, receptors, heparin-binding, phospholipid-binding, integrins, immune response, and cell adhesion molecules, while ectosomes are enriched in endoplasmic reticulum proteasome and mitochondrial proteins [9].

The amounts of different lipid classes in EVs have been determined in several studies [23, 34], and the enrichment of EV membranes for cholesterol, sphingomyelin, glycosphingolipids, and phosphatidylserine compared with their cellular sources was proved. However, differences in lipid composition were reported between vesicle type and cellular source. Generally, exosomes seem to be enriched in glycolipids, phosphatidylserine, and free fatty acids, while ceramides and sphingomyelins were consistently enriched in ectosomes. Still, phosphatidylcholines were depleted in exosomes but unchanged or enriched in ectosomes [9].

EVs contain not only proteins and lipids, but several classes of RNAs. Most of the recent in vitro studies have proved that EVs contain functional RNA molecules that reflect the cellular status and are involved in intercellular crosstalk [18, 19]. Different species of RNA have been reported to be enclosed in EVs derived from various sources—mRNA, rRNA, and tRNA fragments and especially microRNA (miRs) [5]. Several mechanisms for RNA selection, loading into EVs, and their uptake by various target cells have been proposed [5, 20]. Packing into EVs protects the molecules from RNase degradation once released into the extracellular environment. Thus, RNA molecules can be transferred to distant recipient cells, their protein production can be modulated [8, 21], or they may be used as predictive biomarkers for the occurrence of cardiovascular events as demonstrated by the study of EVs containing miR-199a and miR-126 in patients with stable coronary artery disease [22]. In atherosclerotic disease, miR-containing circulating EVs and apoptotic bodies, along with other bioactive molecules, are released by proinflammatory stimulated monocytes and T cells; ECs and activated platelets initiate hyperplasia of vascular smooth muscle cells (VSMCs) which leads to phenotype switching from contractile to synthetic and activates their proliferation and migration [23].

Besides membrane proteins and RNA cargo, EVs may contain cytosolic proteins, such as cytokines, chemokines, growth factors, enzymes or transcription factors, functional organelles, and other bioactive molecules such as lipid mediators, derived from arachidonic acid [5]. Also, some EVs seem to retain the capacity to synthesize eicosanoids using their phospholipid content both by enzymatic and nonenzymatic processes [24].

All neural cells from rodent [25, 26] and human [25, 27], even immortalized human brain microvascular ECs [25, 28], release EVs which contain mRNA and miRs for epigenetic reprogramming of neural cells or post-transcriptional control of specific genes [25]. In vitro studies of brain angiogenesis revealed that EVs deliver proangiogenic protein, mRNAs, and miRs from cultured glioblastoma cells into cerebral ECs [25, 29], especially increased VEGFR-B from immortalized mouse cerebral ECs stimulated with LPS and cytokines into targeted cerebral vascular pericytes [25, 30]. In vitro and in vivo studies showed that neuronal exosomes containing miR-132 could mediate neuronal regulation of brain vascular integrity. Thus, in zebrafish larvae and cultured rodent brain cells, it has been shown that neurons transfer miR-132, a highly conserved and neuron-enriched miR, via secreting exosomes to ECs to maintain brain vascular integrity. Following translocation to ECs through exosome internalization, miR-132 regulates the expression of vascular ECs cadherin (VE-cadherin), an important adherens junction protein, by directly targeting eukaryotic elongation factor 2 kinase [31]. In addition, two proteins found in peripherally circulating plasma EVs, cystatin C and CD14, have been linked to the development of brain atrophy and to cerebral white matter lesions, a small vessel disease within the brain [32]. Because exosomes contain transferrin and insulin receptor [25, 28], which mediate macromolecular passing through the blood-brain barrier (BBB), peripherally infused modified exosomes containing specific RNA were used to knockdown a specific gene in mouse brain [32, 33, 34]. Considering the extraordinary intricate cytoarchitecture of the brain, the presence of EVs in the adult brain is hard to be documented. Fetal brain and neurospheres contain cells which seem to release vesicles into the extracellular space (Figure 1). EVs are easier to be seen near ependymal cells floating in the ventricles from where they can be isolated [11].

Figure 1.

Transmission electron microscopy of the isolated extracellular vesicles: (A) negative staining and (B) cryo-electron microscopy.

Advertisement

3. Methods of isolation and analysis of extracellular vesicles

Minimal experimental requirements for definition of EVs and their functions have been proposed [35], and isolated EVs should be characterized, and the morphology, protein composition, and functionality should be tested before any new enquiries are pursued. Whether the chosen isolation technique for the experiment consists in ultracentrifugation or any other technique, after collecting the sample it is necessary to perform two low-speed centrifugations as soon as possible after the sample collection [36]. The first low-speed centrifugation (300–800×g) removes cells, lipid droplets, and macromolecules from the sample, while the second low-speed centrifugation (2.500×g) removes platelets and apoptotic bodies. Cell removal is an important step; otherwise, the following high-speed centrifugations break the cells, leading to EV release and sample contamination [37].

Differential ultracentrifugation is the most widely used method for EV isolation [36], which involves multiple centrifugations at increasingly higher speeds obtaining a particle separation based on the sedimentation coefficient [38]. Establishing the appropriate speed and duration of centrifugation is a very important step in EV isolation [36]. Several inconveniences can occur using this technique such as the loss of certain subpopulations of EVs or simultaneous isolation of lipo/protein aggregates [39, 40].

Density gradient ultracentrifugation is a method that isolates the particles by size and mass density [41], and its usefulness lies in the fact that it can isolate several subpopulations of EVs which are lost using differential ultracentrifugation; therefore, it increases the purity of the isolated EVs [42].

Size exclusion chromatography is an easy handling method by which a size-based separation is achieved [43]. It allows the electron microscopy to be performed immediately after isolation and the proteins and lipoproteins are removed from the sample without losing any subpopulation, thus achieving a high-yield separation [44].

Precipitation kits gained a lot of attention during the last years involving a concentration method based on the use of polyethylene glycol polymers. The price for the kits is low, the protocol is easy to perform, it is compatible with both low and high volumes of samples, and the method can be applied on large scale [45]. It must be taken into account that the precipitation kits are not the best method for EV isolation, having a low-yield purification because of the co-precipitations [37]. Anyway, for validating the results, it is recommended to use one of the ultracentrifugation methods in parallel with this method.

After the isolation step, a general description of the protein composition should be made even though there are no specific EV markers discovered yet. EVs are enriched in certain proteins such as tetraspanins (CD63, CD81, and CD9) and TSG 101 for exosomes, annexin V for ectosomes, etc. [46]. To exclude a contamination, the presence or absence of proteins that are not expected to be found in EVs, intracellular proteins like calnexin, cytochrome C, histones, GRP94, and Argonaute complex must be detected [35]. Western blot, flow cytometry, or mass spectroscopy can be used for this protein characterization step.

The next step consists in individual characterization of the EVs using at least two methods: electron microscopy [35, 45], atomic force microscopy [47], or nanoparticle tracking analysis [48]. Transmission electron microscopy (TEM) is the most used method for visualization of EVs considering their nanoscale size (Figures 1 and 2). Negative staining [49], TEM on thin section from plastic embedded EVs [49], can also be used to confirm the presence of the lipid bilayer of the isolated EV membrane and an election method to investigate EVs in tissue. Cryo-TEM [50] is a high resolution method to visualize the lipid bilayer. Electron tomography [4, 51] allows 3D reconstruction of the EVs with a very good imaging of the different types. For surface protein recognition, immuno-EM [52, 53] allows imaging of EV-specific markers.

Figure 2.

Transmission electron microscopy: (A) ependymal cells (EC) facing the ventricular system of the mouse brain control the communication between cerebrospinal fluid and brain parenchyma; C—capillary. (B) Higher magnification of the square-marked area in (A) shows extracellular vesicles (arrows) close to cilia and microvilli of the ependymal cell (EC). Volume transmission may be mediated by multivesicular cargoes (encircled) which could be seen released from neurons in fetal mouse brain (C) or neurons from neurospheres generated from stem cells (D).

Flow cytometry is a method based on measuring the signal of the light scattering from a structure passing through a laser beam. The smaller the particles, the less the light scattering, and therefore, conventional cytometry has a detection limit of approximately 200 nm [54]. Fluorescence-based detection is used to overcome this inconvenience [45, 55], the background noise being higher than the EV signal.

Nanoparticle tracking analysis involves the passing of a laser beam through a suspension that contains EVs and the scattering light is captured by a camera [48]. The Brownian movement of the EVs in the suspension is recorded, and according to their movement, the size can be measured [48].

The last step to fulfill the minimal experimental requirements involves the functional assays [35]. Multiple in vitro models can be designed for this step; co-incubating EVs with different culture cells and migration, proliferation, coagulation, and fibrinolysis may be quantified depending on the chosen model [45].

There is no gold-standard method yet, but researchers are encouraged to explore and to standardize their methods of isolation with the hope that someday, easy EV isolation will be possible.

Advertisement

4. Extracellular vesicles and cardiovascular risk for brain pathology

All cells are capable of secreting specific arrays of EVs. Furthermore, the same cells may produce EVs by different biogenesis pathways, with different intracellular origins, of various sizes, with diverse composition and consequently, different functional properties [17]. Crucial feature for the normal functionality of the CNS and brain microenvironment homeostasis is sustained by vascular integrity which is highly dependent on the systemic status.

Several studies of patients with stable coronary artery disease have reported increased levels of circulating EVs which may influence the BBB integrity. Specific EV subpopulations, especially those of ECs origin, characterized by CD144+, CD131+/annexin A5+, or EVs containing miR-199a and miR-126, are currently researched as interesting biomarkers for cardiovascular risk and mortality in these patients [22, 56, 57].

Calcifications present in the atherosclerotic plaque [58] have a destabilizing effect in early lesions, favoring the rupture, but gain a potential protective effect in advanced lesions with heavy calcium deposits [59]. As several studies have shown, atherosclerotic plaque calcifications are associated with EVs of ECs, VSMCs, and macrophage origin. VSMCs can release EVs with low levels of calcification-inhibiting enzymes and annexin A6/phosphatidylserine nucleation complexes. Exposure of VSMCs to proinflammatory cytokines can stimulate the release of EVs that can mineralize when inhibitors of calcification are missing [60]. Also, ECs exposed to proinflammatory stimuli can release EVs rich in bone morphogenetic protein 2, promoting calcification in VSMs [61]. Alterations in local homeostasis of calcium and phosphate lead to the formation of macrophage-derived exosomes which stimulate mineralization, through an annexin-dependent mechanism [62].

Recent studies have also noted that in humans, advanced atherosclerotic plaques have a high content of procoagulant EVs, originating form leukocytes, erythrocytes, and VSMCs [63, 64]. In opposition to ectosomes, exosomes have shown antithrombotic effects. In animal studies, platelet-derived exosomes suppressed platelet aggregation and occlusive thrombosis by inhibiting platelet CD36 and decreasing CD36-dependent oxidized LDL binding and macrophage cholesterol loading [65]. EVs influence the different mechanisms that lead to plaque destabilization and rupture. Intraplaque hemorrhages are produced by neovascularization originating from adventitial tissue, stimulated by plaque EVs, such as CD40+ vesicles of macrophage origin. Hemorrhages are also favored by leukocyte and ECs-EVs with fibrinolytic activity [66, 67]. Fibrous cap weakening is associated with VSMC apoptosis, induced by the presence of EVs, released in some pathological conditions [100]. Moreover, EVs can influence breakdown of matrix structural proteins through metalloproteinase interactions [68].

Circulating levels of procoagulant EVs are higher in patients with acute coronary syndromes compared to healthy controls or patients with stable coronary artery disease [57, 69]. Circulating EVs alter endothelial-dependent NO mediated vasodilation, and endothelial EVs increase ECs thrombogenicity [70, 71]. Circulating EVs have also been investigated as prognostic markers in secondary prevention, in order to identify patients at high cardiovascular risk [56, 57]. Increased levels of CD11b+/CD66+ leukocyte-derived EVs could be useful in identifying asymptomatic patients at high risk for plaque rupture [72], while CD3+/CD45+ EVs could identify individuals who will develop a major cardiovascular event [73].

All these circulating EVs associated with coronary disease [74] may affect the vascular bed of the brain and disrupt the functionality of BBB. Aging and cardiovascular-associated disease are associated with BBB alteration [75] and blood circulating EVs may mediate early dysfunctions or progression of cerebral associated pathology. EVs associated with hypercholesterolemia [76] and atherosclerosis [77, 78] may have an impact on BBB function, and reducing the proinflammatory cytokines enrolling in EVs [79] may be beneficial.

Different neuronal cell types and molecules concur to regulate the improvement of brain vasculature [31, 80, 81, 82]. Each cell, including neurons and astrocytes [32, 83], is able to produce EVs enriched in specific proteins, lipids, and RNAs. EVs can stimulate targeted neural cells and surrounding neural tissues, which are important elements of vascular integrity preservation. The brain pathological condition changes the EV content profile of proteins and miRs [84]. A number of studies have shown that EVs regulate arterial stiffness [32, 85] which is linked to small vessel [32, 86, 87, 88], and platelet-derived EVs seem to be important players in the formation of cerebral microthrombi which lead to brain atrophy and consequent cognitive degeneration. It is believed that the prothrombotic nature of an elevated number of platelet-derived EVs reported in the acute phase of cerebral infarction may conduct to infarct progression [32, 84, 89, 90]. EV proteins cystatin C and CD14 have been shown to be related to cerebral white matter lesions and the progression of brain atrophy in patients with manifest vascular disease, suggesting a role for EVs in the etiology of structural brain changes [32].

The role of cardiovascular disease risk factors in the occurrence and progression of cognitive impairment is widely accepted. There is a link between elevated levels of cholesterol and amyloid deposition in the brain, and the relationship between atherosclerotic injury and sporadic Alzheimer’s disease is investigated.

Advertisement

5. Extracellular vesicles in neurodegenerative diseases

The pathology of Alzheimer’s disease (AD) consists in the extracellular amyloid plaques formed by aggregated amyloid beta peptides and in the intraneuronal neurofibrillary tangles made of hyperphosphorylated tau proteins [91]. The accumulation of the proteins induces an apoptotic response with neuronal loss and occurs especially in the cerebral cortex [92].

It has been observed that the pathological findings in AD have a typical spatial distribution suggesting a neuron-to-neuron spread of the amyloid and hyperphosphorylated tau proteins, which promote aggregation, acting as “seeds” [93]. Therefore AD is considered to have a prion-like model of propagation [94]. The immediate question that rises is: what is the mechanism of the propagation?

Amyloid beta is mainly formed extracellularly from the cleavage of APP by beta and gamma secretases, which are found at the level of the plasma membrane [95]. But in some degree, the secretases are present in endoplasmic reticulum and Golgi apparatus, and there is a certain intracellular production of amyloid beta [96]. It is removed from the cell via exosomes embedded in a multivesicular body as an alternative pathway to lysosomal degradation [97]. A new study suggests that exosomes containing amyloid beta are present in higher concentrations in the AD brain compared to the healthy brain [98]. Moreover, the study shows that exosomes are the carriers of the toxic amyloid beta from one neuron to another [98]. Also, it is thought that exosomes mediate the intercellular transfer of hyperphosphorylated tau protein, and the exosome-mediated tau protein induces the formation of neurofibrillary tangles [99].

The evolution of AD is insidious with an asymptomatic stage that lasts several years [100]. Although asymptomatic, the pathological changes in the brain are present in this stage [93]. These findings suggest the value of discovering biomarkers that can anticipate the onset of the clinical symptoms or can facilitate a window for the possibility of a future therapy that could stop the progression. Amyloid beta and hyperphosphorylated tau proteins are of great value as biomarkers when dosed from the cerebrospinal fluid [101]. Nevertheless, performing lumbar puncture in a wide population is almost impossible. Thereby, the discovery of new biomarkers is a valuable research theme.

Several types of miRs isolated from cerebrospinal fluid are differentially expressed in AD, such as miR-100, miR-146, miR-505, and miR-1274a [102]. The presence of several types of exosomal miRs isolated from serum (miR-361-5p, miR-93-5p, miR-335-5p, and miR-30e-5p) correlates with the neuropsychological evaluation and brain imaging [103]. It is to be mentioned that exosome-containing proteins like tau, apolipoprotein E, cystatin E, and HSP-90 alpha were isolated in the cerebrospinal fluid and were present in patients with AD [104]. This evidence proposes both miRs and protein-containing exosomes as a promising source of biomarkers for AD.

Parkinson’s disease (PD) is a neurodegenerative disease that consists in the loss of dopaminergic neurons localized in the substantia nigra [105]. The pathology of the disease implies the deposition of Lewy bodies in the neurons which are mostly made of misfolded and aggregated alpha-synuclein protein [105]. The Braak staging explains the spatio-temporal dissemination of Lewy bodies into the neurons from caudal to rostral, starting in the medulla oblongata and spreading to the level of the cerebral cortex, damaging various structures on this way [106]. The starting point is thought to be either the enteric nervous system or the olfactory bulb [106]. Thereby, it is suggested that PD has a prion-like propagation [94]. Are exosomes responsible for carrying alpha-synuclein from neuron to neuron? The mechanism could be similar to the one described in AD, but there are fewer studies to draw certain conclusions regarding the involvement of exosomes in the pathogenesis of PD. Excessive intracellular alpha-synuclein is thought to be transported out of the cell via multivesicular body containing exosomes in a similar manner as in AD in an attempt to clean the intracellular space [107].

Studies of neuronal cell cultures expressing alpha-synuclein noticed that the protein is released by the cell free in the extracellular space but also incorporated in the exosomes [108]. Furthermore, neighboring cells take up the exosomes, and the transferred alpha-synuclein acts as seed for the formation of the aggregate [109]. Evidence to prove that in vivo alpha-synuclein is transferred by exosomes is not available yet, but it can be speculated that exosomes play an important role as in vitro.

The use of EVs as a biomarker tool in PD is a promising field too [110]. Several miRs carried by exosomes were increased in the CSF of PD patients, such as miR-10a-5p, miR-153, and miR-409-3p, while some miRs such as miR-19b-3p and miR-1 were significantly reduced [111]. Exosomes containing alpha-synuclein and LRRK proteins were also isolated in the CSF of PD patients [109, 112].

Advertisement

6. Extracellular vesicles’ perspective use in brain pathology

The discovery of the EV involvement in several biological processes gave hope that some questions regarding neurodegenerative diseases will be answered. First of all, it is important to clarify which are the cellular mechanisms involved in the progression of the disease and if exosomes play any role. Based on the spatio-temporal spreading of the pathological proteins in the brain, an appealing theory is the prion-like propagation theory [113]. It is presumed that exosomes play an important role by facilitating the interneuronal transport of the proteins [114]. As well, there is a critical need in finding accessible biomarkers that can diagnose a neurodegenerative disease in the asymptomatic stage [115]. Dosing certain free proteins in biofluids can be an option, but several problems are experienced because of their low concentrations [115]. Therefore, a new approach is being attempted consisting in finding the proteins encapsulated in extracellular vesicles [116]. Micro-RNAs and different proteins carried by exosomes are attractive options for finding new biomarkers in several diseases as well as in neurodegenerative diseases [117].

Recently, it was discovered that an important player in the field of neural diseases is EV derived from stem cells [118], mainly in the case of stroke [118, 119, 120]. In stroke pathophysiology, inflammation plays a significant role, circulating EV-activating immune cells. Neurons quickly depolarize and die, being next phagocytosed by infiltrating circulating macrophages and microglia [84, 121].

Because of their β1 and α2b integrin-enriched content, human neural stem cell-derived EV administration recovers both tissue and sensorimotor function and may protect the BBB integrity, in the preclinical mouse thromboembolic model of stroke [122, 123]. Similarly, multipotent mesenchymal stromal cells (MSCs), through their capacity to secrete soluble factors, play an important role in brain repair. It was demonstrated that MSC cargos modulate cell signaling in ischemic stroke by PI3K/Akt pathway activation [123, 124] and EVs facilitated secretion of miRs sustaining MSC neuroprotective effects in ischemic stroke as well. Previous research demonstrated that intravenous administration of bone marrow-MSCs containing exosomes transferred miR-133b to astrocytes and neurons into the ischemic boundary zone [120, 123], and MSCs cultured in the presence of extracts from rat ischemic brain induced increased expression of exosomal miR133b [123, 125]. Also, EVs released by human MSCs seem to have an anti-inflammatory effect on mast cells, by increased prostaglandin E2 (PGE2) synthesis and up-regulation of EP4 receptor which might prevent the rupture of intracranial aneurysms [126]. All these data suggest that EVs from various sources may contribute to the neurogenesis and angiogenesis during brain repair processes in cerebral diseases.

Advertisement

7. Conclusion

The EVs emerge as a powerful tool for early diagnosis and subsequent prevention of pathologies with high risk for the brain dysfunction. Still, EVs investigation as biomarkers and therapeutic agents is in its infancy. There is increasing evidence that EVs have an important role in brain pathophysiology. Thus, their potential application as prognostic and diagnostic biomarkers and targeted therapeutic tools relies on their subsequent isolation for molecular and functional characterization that can relate them with the cellular source and cellular mechanisms in both health and disease.

Advertisement

Acknowledgments

This work was supported by grants of UEFISCDI, projects no. PN-III-P1-1.2-PCCDI-2017-0527 and PN-III-P1-1.2-PCCDI-2017-0797 and the Romanian Academy and Romanian Ministry of Research and Innovation, projects no. 1N/2019_19.29.01.02 and 7PFE/16.10.2018.

Advertisement

Conflict of interest

The authors declare that the research was conducted in the absence of any either commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. 1. Gould SJ, Raposo G. As we wait: Coping with an imperfect nomenclature for extracellular vesicles. Journal of Extracellular Vesicles. 2013;2:20389. DOI: 10.3402/jev.v2i0.20389
  2. 2. Cocucci E, Meldolesi J. Ectosomes and exosomes: Shedding the confusion between extracellular vesicles. Trends in Cell Biology. 2015;25:364-372. DOI: 10.1016/j.tcb.2015.01.004
  3. 3. van der Pol E, Böing AN, Gool EL, Nieuwland R. Recent developments in the nomenclature, presence, isolation, detection and clinical impact of extracellular vesicles. Journal of Thrombosis and Haemostasis. 2016;14:48-56. DOI: 10.1111/jth.13190
  4. 4. Fertig ET, Gherghiceanu M, Popescu LM. Extracellular vesicles release by cardiac telocytes: Electron microscopy and electron tomography. Journal of Cellular and Molecular Medicine. 2014;18:1938-1943. DOI: 10.1111/jcmm.12436
  5. 5. Abels ER, Breakefield XO. Introduction to extracellular vesicles: Biogenesis, RNA cargo selection, content, release, and uptake. Cellular and Molecular Neurobiology. 2016;36:301-312. DOI: 10.1007/s10571-016-0366-z
  6. 6. Maas SLN, Breakefield XO, Weaver AM. Extracellular vesicles: Unique intercellular delivery vehicles. Trends in Cell Biology. 2017;27:172-188. DOI: 10.1016/j.tcb.2016.11.003
  7. 7. van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nature Reviews. Molecular Cell Biology. 2018;19:213-228. DOI: 10.1038/nrm.2017.125
  8. 8. Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature Cell Biology. 2007;9:654-659. DOI: 10.1038/ncb1596
  9. 9. Haraszti RA, Didiot M-C, Sapp E, Leszyk J, Shaffer SA, Rockwell HE, et al. High-resolution proteomic and lipidomic analysis of exosomes and microvesicles from different cell sources. Journal of Extracellular Vesicles. 2016;5:32570. DOI: 10.3402/jev.v5.32570
  10. 10. Thompson AG, Gray E, Mager I, Fischer R, Thézénas M, Charles PD, et al. UFLC-derived CSF extracellular vesicle origin and proteome. Proteomics. 2018;18:1800257. DOI: 10.1002/pmic.201800257
  11. 11. Welton JL, Loveless S, Stone T, von Ruhland C, Robertson NP, Clayton A. Cerebrospinal fluid extracellular vesicle enrichment for protein biomarker discovery in neurological disease; multiple sclerosis. Journal of Extracellular Vesicles. 2017;6:1369805. DOI: 10.1080/20013078.2017.1369805
  12. 12. Pulliam L, Sun B, Mustapic M, Chawla S, Kapogiannis D. Plasma neuronal exosomes serve as biomarkers of cognitive impairment in HIV infection and Alzheimer’s disease. Journal of Neurovirology. 2019. DOI: 10.1007/s13365-018-0695-4. [Epub ahead of print]
  13. 13. Georgescu A, Alexandru N, Popov D, Amuzescu M, Andrei E, Zamfir C, et al. Chronic venous insufficiency is associated with elevated level of circulating microparticles. Journal of Thrombosis and Haemostasis. 2009;7:1566-1575. DOI: 10.1111/j.1538-7836.2009.03525.x
  14. 14. Pardo F, Villalobos-Labra R, Sobrevia B, Toledo F, Sobrevia L. Extracellular vesicles in obesity and diabetes mellitus. Molecular Aspects of Medicine. 2018;60:81-91. DOI: 10.1016/j.mam.2017.11.010
  15. 15. French KC, Antonyak MA, Cerione RA. Extracellular vesicle docking at the cellular port: Extracellular vesicle binding and uptake. Seminars in Cell & Developmental Biology. 2017;67:48-55. DOI: 10.1016/j.semcdb.2017.01.002
  16. 16. Tkach M, Théry C. Communication by extracellular vesicles: Where we are and where we need to go. Cell. 2016;164:1226-1232. DOI: 10.1016/j.cell.2016.01.043
  17. 17. Kowal J, Arras G, Colombo M, Jouve M, Morath JP, Primdal-Bengtson B, et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proceedings of the National Academy of Sciences of the United States of America. 2016;113:E968-E977. DOI: 10.1073/pnas.1521230113
  18. 18. de Jong OG, Verhaar MC, Chen Y, Vader P, Gremmels H, Posthuma G, et al. Cellular stress conditions are reflected in the protein and RNA content of endothelial cell-derived exosomes. Journal of Extracellular Vesicles. 2012;1:18396. DOI: 10.3402/jev.v1i0.18396
  19. 19. Pérez-Boza J, Lion M, Struman I. Exploring the RNA landscape of endothelial exosomes. RNA. 2018;24:423-435. DOI: 10.1261/rna.064352.117
  20. 20. Frankel EB, Audhya A. ESCRT-dependent cargo sorting at multivesicular endosomes. Seminars in Cell & Developmental Biology. 2018;74:4-10. DOI: 10.1016/j.semcdb.2017.08.020
  21. 21. Njock M-S, Cheng HS, Dang LT, Nazari-Jahantigh M, Lau AC, Boudreau E, et al. Endothelial cells suppress monocyte activation through secretion of extracellular vesicles containing anti-inflammatory microRNAs. Blood. 2015;125:3202-3212. DOI: 10.1182/blood-2014-11-611046
  22. 22. Jansen F, Yang X, Proebsting S, Hoelscher M, Przybilla D, Baumann K, et al. MicroRNA expression in circulating microvesicles predicts cardiovascular events in patients with coronary artery disease. Journal of the American Heart Association. 2014;3:e001249. DOI: 10.1161/JAHA.114.001249
  23. 23. Chistiakov DA, Orekhov AN, Bobryshev YV. Extracellular vesicles and atherosclerotic disease. Cellular and Molecular Life Sciences. 2015;72:2697-2708. DOI: 10.1007/s00018-015-1906-2
  24. 24. Boilard E. Extracellular vesicles and their content in bioactive lipid mediators: More than a sack of microRNA. Journal of Lipid Research. 2018;59:2037-2046. DOI: 10.1194/jlr.R084640
  25. 25. Zhang ZG, Chopp M. Exosomes in stroke pathogenesis and therapy. The Journal of Clinical Investigation. 2016;126:1190-1197. DOI: 10.1172/JCI81133
  26. 26. Perez-Gonzalez R, Gauthier SA, Kumar A, Levy E. The exosome secretory pathway transports amyloid precursor protein carboxyl-terminal fragments from the cell into the brain extracellular space. The Journal of Biological Chemistry. 2012;287:43108-43115. DOI: 10.1074/jbc.M112.404467
  27. 27. Banigan MG, Kao PF, Kozubek JA, Winslow AR, Medina J, Costa J, et al. Differential expression of exosomal microRNAs in prefrontal cortices of schizophrenia and bipolar disorder patients. PLoS One. 2013;8:e48814. DOI: 10.1371/journal.pone.0048814
  28. 28. Haqqani AS, Delaney CE, Tremblay T-L, Sodja C, Sandhu JK, Stanimirovic DB. Method for isolation and molecular characterization of extracellular microvesicles released from brain endothelial cells. Fluids and Barriers of the CNS. 2013;10:4. DOI: 10.1186/2045-8118-10-4
  29. 29. Skog J, Würdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nature Cell Biology. 2008;10:1470-1476. DOI: 10.1038/ncb1800
  30. 30. Yamamoto S, Niida S, Azuma E, Yanagibashi T, Muramatsu M, Huang TT, et al. Inflammation-induced endothelial cell-derived extracellular vesicles modulate the cellular status of pericytes. Scientific Reports. 2015;5:8505. DOI: 10.1038/srep08505
  31. 31. Xu B, Zhang Y, Du X-F, Li J, Zi H-X, Bu J-W, et al. Neurons secrete miR-132-containing exosomes to regulate brain vascular integrity. Cell Research. 2017;27:882-897. DOI: 10.1038/cr.2017.62
  32. 32. Kanhai DA, de Kleijn DPV, Kappelle LJ, Uiterwaal CSPM, van der Graaf Y, Pasterkamp G, et al. Extracellular vesicle protein levels are related to brain atrophy and cerebral white matter lesions in patients with manifest vascular disease: The SMART-MR study. BMJ Open. 2014;4:e003824. DOI: 10.1136/bmjopen-2013-003824
  33. 33. Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJA. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nature Biotechnology. 2011;29:341-345. DOI: 10.1038/nbt.1807
  34. 34. Graner MW, Alzate O, Dechkovskaia AM, Keene JD, Sampson JH, Mitchell DA, et al. Proteomic and immunologic analyses of brain tumor exosomes. The FASEB Journal. 2009;23:1541-1557. DOI: 10.1096/fj.08-122184
  35. 35. Lötvall J, Hill AF, Hochberg F, Buzás EI, Di Vizio D, Gardiner C, et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: A position statement from the International Society for Extracellular Vesicles. Journal of Extracellular Vesicles. 2014;3:26913
  36. 36. Théry C, Amigorena S, Raposo G, Clayton A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Current Protocols in Cell Biology. 2006;30:3.22.1-3.22.29. DOI: 10.1002/0471143030.cb0322s30
  37. 37. Witwer KW, Buzás EI, Bemis LT, Bora A, Lässer C, Lötvall J, et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. Journal of Extracellular Vesicles. 2013;2:20360. DOI: 10.3402/jev.v2i0.20360
  38. 38. Livshits MA, Khomyakova E, Evtushenko EG, Lazarev VN, Kulemin NA, Semina SE, et al. Isolation of exosomes by differential centrifugation: Theoretical analysis of a commonly used protocol. Scientific Reports. 2015;5:17319. DOI: 10.1038/srep17319
  39. 39. Gyorgy B, Modos K, Pallinger E, Paloczi K, Pasztoi M, Misjak P, et al. Detection and isolation of cell-derived microparticles are compromised by protein complexes resulting from shared biophysical parameters. Blood. 2011;117:e39-e48. DOI: 10.1182/blood-2010-09-307595
  40. 40. Yuana Y, Böing AN, Grootemaat AE, van der Pol E, Hau CM, Cizmar P, et al. Handling and storage of human body fluids for analysis of extracellular vesicles. Journal of Extracellular Vesicles. 2015;4:29260. DOI: 10.3402/jev.v4.29260
  41. 41. Greening DW, Xu R, Ji H, Tauro BJ, Simpson RJ. A protocol for exosome isolation and characterization: Evaluation of ultracentrifugation, density-gradient separation, and immunoaffinity capture methods. Methods in Molecular Biology. 2015;1295:179-209. DOI: 10.1007/978-1-4939-2550-6_15
  42. 42. Willms E, Johansson HJ, Mäger I, Lee Y, Blomberg KEM, Sadik M, et al. Cells release subpopulations of exosomes with distinct molecular and biological properties. Scientific Reports. 2016;6:22519. DOI: 10.1038/srep22519
  43. 43. Koh YQ , Almughlliq FB, Vaswani K, Peiris HN, Mitchell MD. Exosome enrichment by ultracentrifugation and size exclusion chromatography. Frontiers in Bioscience (Landmark Ed). 2018;23:865-874
  44. 44. Böing AN, van der Pol E, Grootemaat AE, Coumans FAW, Sturk A, Nieuwland R. Single-step isolation of extracellular vesicles by size-exclusion chromatography. Journal of Extracellular Vesicles. 2014;3:23430. DOI: 10.3402/jev.v3.23430
  45. 45. Coumans FAW, Brisson AR, Buzas EI, Dignat-George F, Drees EEE, El-Andaloussi S, et al. Methodological guidelines to study extracellular vesicles. Circulation Research. 2017;120:1632-1648. DOI: 10.1161/CIRCRESAHA.117.309417
  46. 46. Kalra H, Drummen GPC, Mathivanan S. Focus on extracellular vesicles: Introducing the next small big thing. International Journal of Molecular Sciences. 2016;17:170. DOI: 10.3390/ijms17020170
  47. 47. Yuana Y, Oosterkamp TH, Bahatyrova S, Ashcroft B, Garcia Rodriguez P, Bertina RM, et al. Atomic force microscopy: A novel approach to the detection of nanosized blood microparticles. Journal of Thrombosis and Haemostasis. 2010;8:315-323. DOI: 10.1111/j.1538-7836.2009.03654.x
  48. 48. Gardiner C, Shaw M, Hole P, Smith J, Tannetta D, Redman CW, et al. Measurement of refractive index by nanoparticle tracking analysis reveals heterogeneity in extracellular vesicles. Journal of Extracellular Vesicles. 2014;3:25361. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25425324. [Accessed: 14 Oct 2018]
  49. 49. Jung MK, Mun JY. Sample preparation and imaging of exosomes by transmission electron microscopy. Journal of Visualized Experiments. 2018. DOI: 10.3791/56482
  50. 50. Yuana Y, Koning RI, Kuil ME, Rensen PCN, Koster AJ, Bertina RM, et al. Cryo-electron microscopy of extracellular vesicles in fresh plasma. Journal of Extracellular Vesicles. 2013;2:21494. DOI: 10.3402/jev.v2i0.21494
  51. 51. Coleman BM, Hanssen E, Lawson VA, Hill AF. Prion-infected cells regulate the release of exosomes with distinct ultrastructural features. The FASEB Journal. 2012;26:4160-4173. DOI: 10.1096/fj.11-202077
  52. 52. Cizmar P, Yuana Y. Detection and characterization of extracellular vesicles by transmission and cryo-transmission electron microscopy. Methods in Molecular Biology. 2017;1660:221-232. DOI: 10.1007/978-1-4939-7253-1_18
  53. 53. Arraud N, Linares R, Tan S, Gounou C, Pasquet J-M, Mornet S, et al. Extracellular vesicles from blood plasma: Determination of their morphology, size, phenotype and concentration. Journal of Thrombosis and Haemostasis. 2014;12:614-627. DOI: 10.1111/jth.12554
  54. 54. Nolan JP. Flow cytometry of extracellular vesicles: Potential, pitfalls, and prospects. Current Protocols in Cytometry. 2015;73:13.14.1-16. DOI: 10.1002/0471142956.cy1314s73
  55. 55. van der Vlist EJ, Nolte-‘t Hoen ENM, Stoorvogel W, Arkesteijn GJA, Wauben MHM. Fluorescent labeling of nano-sized vesicles released by cells and subsequent quantitative and qualitative analysis by high-resolution flow cytometry. Nature Protocols. 2012;7:1311-1326. DOI: 10.1038/nprot.2012.065
  56. 56. Sinning J-M, Losch J, Walenta K, Böhm M, Nickenig G, Werner N. Circulating CD31+/Annexin V+ microparticles correlate with cardiovascular outcomes. European Heart Journal. 2011;32:2034-2041. DOI: 10.1093/eurheartj/ehq478
  57. 57. Nozaki T, Sugiyama S, Koga H, Sugamura K, Ohba K, Matsuzawa Y, et al. Significance of a multiple biomarkers strategy including endothelial dysfunction to improve risk stratification for cardiovascular events in patients at high risk for coronary heart disease. Journal of the American College of Cardiology. 2009;54:601-608. DOI: 10.1016/j.jacc.2009.05.022
  58. 58. Aikawa E. Extracellular vesicles in cardiovascular disease: Focus on vascular calcification. The Journal of Physiology. 2016;594:2877-2880. DOI: 10.1113/JP272112
  59. 59. Hsu JJ, Lim J, Tintut Y, Demer LL. Cell-matrix mechanics and pattern formation in inflammatory cardiovascular calcification. Heart. 2016;102:1710-1715. DOI: 10.1136/heartjnl-2016-309667
  60. 60. Kapustin AN, Chatrou MLL, Drozdov I, Zheng Y, Davidson SM, Soong D, et al. Vascular smooth muscle cell calcification is mediated by regulated exosome secretion. Circulation Research. 2015;116:1312-1323. DOI: 10.1161/CIRCRESAHA.116.305012
  61. 61. Buendía P, Montes de Oca A, Madueño JA, Merino A, Martín-Malo A, Aljama P, et al. Endothelial microparticles mediate inflammation-induced vascular calcification. The FASEB Journal. 2015;29:173-181. DOI: 10.1096/fj.14-249706
  62. 62. New SEP, Aikawa E. Role of extracellular vesicles in de novo mineralization: An additional novel mechanism of cardiovascular calcification. Arteriosclerosis, Thrombosis, and Vascular Biology. 2013;33:1753-1758. DOI: 10.1161/ATVBAHA.112.300128
  63. 63. Mallat Z, Hugel B, Ohan J, Lesèche G, Freyssinet JM, Tedgui A. Shed membrane microparticles with procoagulant potential in human atherosclerotic plaques: A role for apoptosis in plaque thrombogenicity. Circulation. 1999;99:348-353
  64. 64. Leroyer AS, Isobe H, Lesèche G, Castier Y, Wassef M, Mallat Z, et al. Cellular origins and thrombogenic activity of microparticles isolated from human atherosclerotic plaques. Journal of the American College of Cardiology. 2007;49:772-777. DOI: 10.1016/j.jacc.2006.10.053
  65. 65. Srikanthan S, Li W, Silverstein RL, McIntyre TM. Exosome poly-ubiquitin inhibits platelet activation, downregulates CD36 and inhibits pro-atherothombotic cellular functions. Journal of Thrombosis and Haemostasis. 2014;12:1906-1917. DOI: 10.1111/jth.12712
  66. 66. Leroyer AS, Rautou P-E, Silvestre J-S, Castier Y, Lesèche G, Devue C, et al. CD40 ligand+ microparticles from human atherosclerotic plaques stimulate endothelial proliferation and angiogenesis. Journal of the American College of Cardiology. 2008;52:1302-1311. DOI: 10.1016/j.jacc.2008.07.032
  67. 67. Lacroix R, Sabatier F, Mialhe A, Basire A, Pannell R, Borghi H, et al. Activation of plasminogen into plasmin at the surface of endothelial microparticles: A mechanism that modulates angiogenic properties of endothelial progenitor cells in vitro. Blood. 2007;110:2432-2439. DOI: 10.1182/blood-2007-02-069997
  68. 68. Boulanger CM, Loyer X, Rautou P-E, Amabile N. Extracellular vesicles in coronary artery disease. Nature Reviews. Cardiology. 2017;14:259-272. DOI: 10.1038/nrcardio.2017.7
  69. 69. Bernal-Mizrachi L, Jy W, Jimenez JJ, Pastor J, Mauro LM, Horstman LL, et al. High levels of circulating endothelial microparticles in patients with acute coronary syndromes. American Heart Journal. 2003;145:962-970. DOI: 10.1016/S0002-8703(03)00103-0
  70. 70. Boulanger CM, Scoazec A, Ebrahimian T, Henry P, Mathieu E, Tedgui A, et al. Circulating microparticles from patients with myocardial infarction cause endothelial dysfunction. Circulation. 2001;104:2649-2652
  71. 71. Abbas M, Jesel L, Auger C, Amoura L, Messas N, Manin G, et al. Endothelial microparticles from acute coronary syndrome patients induce premature coronary artery endothelial cell aging and thrombogenicity: Role of the Ang II/AT1 receptor/NADPH oxidase-mediated activation of MAPKs and PI3-kinase pathways. Circulation. 2017;135:280-296. DOI: 10.1161/CIRCULATIONAHA.116.017513
  72. 72. Sarlon-Bartoli G, Bennis Y, Lacroix R, Piercecchi-Marti MD, Bartoli MA, Arnaud L, et al. Plasmatic level of leukocyte-derived microparticles is associated with unstable plaque in asymptomatic patients with high-grade carotid stenosis. Journal of the American College of Cardiology. 2013;62:1436-1441. DOI: 10.1016/j.jacc.2013.03.078
  73. 73. Chiva-Blanch G, Suades R, Crespo J, Vilahur G, Arderiu G, Padró T, et al. CD3(+)/CD45(+) and SMA-α(+) circulating microparticles are increased in individuals at high cardiovascular risk who will develop a major cardiovascular event. International Journal of Cardiology. 2016;208:147-149. DOI: 10.1016/j.ijcard.2016.01.211
  74. 74. Alexandru N, Costa A, Constantin A, Cochior D, Georgescu A. Microparticles: From biogenesis to biomarkers and diagnostic tools in cardiovascular disease. Current Stem Cell Research & Therapy. 2017;12:89-102. DOI: 10.2174/1574888X11666151203224058
  75. 75. Ceafalan LC, Fertig TE, Gheorghe TC, Hinescu ME, Popescu BO, Pahnke J, et al. Age-related ultrastructural changes of the basement membrane in the mouse blood-brain barrier. Journal of Cellular and Molecular Medicine. 2019;23:819-827. DOI: 10.1111/jcmm.13980
  76. 76. Alexandru N, Popov D, Dragan E, Andrei E, Georgescu A. Circulating endothelial progenitor cell and platelet microparticle impact on platelet activation in hypertension associated with hypercholesterolemia. PLoS One. 2013;8:e52058. DOI: 10.1371/journal.pone.0052058
  77. 77. Alexandru N, Andrei E, Niculescu L, Dragan E, Ristoiu V, Georgescu A. Microparticles of healthy origins improve endothelial progenitor cell dysfunction via microRNA transfer in an atherosclerotic hamster model. Acta Physiologica (Oxford, England). 2017;221:230-249. DOI: 10.1111/apha.12896
  78. 78. Georgescu A, Alexandru N, Andrei E, Dragan E, Cochior D, Dias S. Effects of transplanted circulating endothelial progenitor cells and platelet microparticles in atherosclerosis development. Biology of the Cell. 2016;108:219-243. DOI: 10.1111/boc.201500104
  79. 79. Georgescu A, Alexandru N, Nemecz M, Titorencu I, Popov D. Irbesartan administration therapeutically influences circulating endothelial progenitor cell and microparticle mobilization by involvement of pro-inflammatory cytokines. European Journal of Pharmacology. 2013;711:27-35. DOI: 10.1016/j.ejphar.2013.04.004
  80. 80. Armulik A, Genové G, Mäe M, Nisancioglu MH, Wallgard E, Niaudet C, et al. Pericytes regulate the blood–brain barrier. Nature. 2010;468:557-561. DOI: 10.1038/nature09522
  81. 81. Daneman R, Zhou L, Kebede AA, Barres BA. Pericytes are required for blood-brain barrier integrity during embryogenesis. Nature. 2010;468:562-566. DOI: 10.1038/nature09513
  82. 82. Argaw AT, Asp L, Zhang J, Navrazhina K, Pham T, Mariani JN, et al. Astrocyte-derived VEGF-A drives blood-brain barrier disruption in CNS inflammatory disease. The Journal of Clinical Investigation. 2012;122:2454-2468. DOI: 10.1172/JCI60842
  83. 83. Fauré J, Lachenal G, Court M, Hirrlinger J, Chatellard-Causse C, Blot B, et al. Exosomes are released by cultured cortical neurones. Molecular and Cellular Neurosciences. 2006;31:642-648. DOI: 10.1016/j.mcn.2005.12.003
  84. 84. Couch Y, Akbar N, Davis S, Fischer R, Dickens AM, Neuhaus AA, et al. Inflammatory stroke extracellular vesicles induce macrophage activation. Stroke. 2017;48:2292-2296. DOI: 10.1161/STROKEAHA.117.017236
  85. 85. Amabile N, Guérin AP, Leroyer A, Mallat Z, Nguyen C, Boddaert J, et al. Circulating endothelial microparticles are associated with vascular dysfunction in patients with end-stage renal failure. Journal of the American Society of Nephrology. 2005;16:3381-3388. DOI: 10.1681/ASN.2005050535
  86. 86. O’Rourke MF, Safar ME. Relationship between aortic stiffening and microvascular disease in brain and kidney: Cause and logic of therapy. Hypertension (Dallas, Tex 1979). 2005;46:200-204. DOI: 10.1161/01.HYP.0000168052.00426.65
  87. 87. Ohmine T, Miwa Y, Yao H, Yuzuriha T, Takashima Y, Uchino A, et al. Association between arterial stiffness and cerebral white matter lesions in community-dwelling elderly subjects. Hypertension Research. 2008;31:75-81. DOI: 10.1291/hypres.31.75
  88. 88. Kuo H-K, Chen C-Y, Liu H-M, Yen C-J, Chang K-J, Chang C-C, et al. Metabolic risks, white matter hyperintensities, and arterial stiffness in high-functioning healthy adults. International Journal of Cardiology. 2010;143:184-191. DOI: 10.1016/j.ijcard.2009.02.005
  89. 89. Cherian P, Hankey GJ, Eikelboom JW, Thom J, Baker RI, McQuillan A, et al. Endothelial and platelet activation in acute ischemic stroke and its etiological subtypes. Stroke. 2003;34:2132-2137. DOI: 10.1161/01.STR.0000086466.32421.F4
  90. 90. Jimenez JJ, Jy W, Mauro LM, Horstman LL, Ahn YS. Elevated endothelial microparticles in thrombotic thrombocytopenic purpura: Findings from brain and renal microvascular cell culture and patients with active disease. British Journal of Haematology. 2001;112:81-90. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11167788. [Accessed: 30 Mar 2019]
  91. 91. Gomez-Isla T, Spires T, De Calignon A, Hyman BT. Neuropathology of Alzheimer’s disease. Handbook of Clinical Neurology. 2008;89:233-243. DOI: 10.1016/S0072-9752(07)01222-5
  92. 92. Cai Z, Zhao B, Ratka A. Oxidative stress and β-amyloid protein in Alzheimer’s disease. Neuromolecular Medicine. 2011;13:223-250. DOI: 10.1007/s12017-011-8155-9
  93. 93. Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathologica. 1991;82:239-259
  94. 94. Duyckaerts C, Clavaguera F, Potier M-C. The prion-like propagation hypothesis in Alzheimerʼs and Parkinsonʼs disease. Current Opinion in Neurology. 2019;32:266-271. DOI: 10.1097/WCO.0000000000000672
  95. 95. Gouras GK, Tampellini D, Takahashi RH, Capetillo-Zarate E. Intraneuronal beta-amyloid accumulation and synapse pathology in Alzheimer’s disease. Acta Neuropathologica. 2010;119:523-541. DOI: 10.1007/s00401-010-0679-9
  96. 96. Hartmann T, Bieger SC, Brühl B, Tienari PJ, Ida N, Allsop D, et al. Distinct sites of intracellular production for Alzheimer’s disease A beta40/42 amyloid peptides. Nature Medicine. 1997;3:1016-1020
  97. 97. Takahashi RH, Milner TA, Li F, Nam EE, Edgar MA, Yamaguchi H, et al. Intraneuronal Alzheimer abeta42 accumulates in multivesicular bodies and is associated with synaptic pathology. The American Journal of Pathology. 2002;161:1869-1879
  98. 98. Sinha MS, Ansell-Schultz A, Civitelli L, Hildesjö C, Larsson M, Lannfelt L, et al. Alzheimer’s disease pathology propagation by exosomes containing toxic amyloid-beta oligomers. Acta Neuropathologica. 2018;136:41. DOI: 10.1007/S00401-018-1868-1
  99. 99. Saman S, Kim W, Raya M, Visnick Y, Miro S, Saman S, et al. Exosome-associated tau is secreted in tauopathy models and is selectively phosphorylated in cerebrospinal fluid in early Alzheimer disease. The Journal of Biological Chemistry. 2012;287:3842-3849. DOI: 10.1074/jbc.M111.277061
  100. 100. Perl DP. Neuropathology of Alzheimer’s disease. Mount Sinai Journal of Medicine A Journal of Translational and Personalized Medicine. 2010;77:32-42. DOI: 10.1002/msj.20157
  101. 101. Tapiola T, Alafuzoff I, Herukka S-K, Parkkinen L, Hartikainen P, Soininen H, et al. Cerebrospinal fluid β-amyloid 42 and tau proteins as biomarkers of Alzheimer-type pathologic changes in the brain. Archives of Neurology. 2009;66. DOI: 10.1001/archneurol.2008.596
  102. 102. Denk J, Boelmans K, Siegismund C, Lassner D, Arlt S, Jahn H. MicroRNA profiling of CSF reveals potential biomarkers to detect Alzheimer’s disease. PLoS One. 2015;10:e0126423. DOI: 10.1371/journal.pone.0126423
  103. 103. Cheng L, Doecke JD, Sharples RA, Villemagne VL, Fowler CJ, Rembach A, et al. Prognostic serum miRNA biomarkers associated with Alzheimer’s disease shows concordance with neuropsychological and neuroimaging assessment. Molecular Psychiatry. 2015;20:1188-1196. DOI: 10.1038/mp.2014.127
  104. 104. Chiasserini D, van Weering JRT, Piersma SR, Pham TV, Malekzadeh A, Teunissen CE, et al. Proteomic analysis of cerebrospinal fluid extracellular vesicles: A comprehensive dataset. Journal of Proteomics. 2014;106:191-204. DOI: 10.1016/j.jprot.2014.04.028
  105. 105. Dickson DW. Neuropathology of Parkinson disease. Parkinsonism & Related Disorders. 2018;46:S30-S33. DOI: 10.1016/j.parkreldis.2017.07.033
  106. 106. Braak H, Del Tredici K, Rüb U, de Vos RAI, Jansen Steur ENH, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiology of Aging. 2013;24:197-211
  107. 107. Danzer KM, Kranich LR, Ruf WP, Cagsal-Getkin O, Winslow AR, Zhu L, et al. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Molecular Neurodegeneration. 2012;7:42. DOI: 10.1186/1750-1326-7-42
  108. 108. Emmanouilidou E, Melachroinou K, Roumeliotis T, Garbis SD, Ntzouni M, Margaritis LH, et al. Cell-produced -synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. The Journal of Neuroscience. 2010;30:6838-6851. DOI: 10.1523/JNEUROSCI.5699-09.2010
  109. 109. Stuendl A, Kunadt M, Kruse N, Bartels C, Moebius W, Danzer KM, et al. Induction of α-synuclein aggregate formation by CSF exosomes from patients with Parkinson’s disease and dementia with Lewy bodies. Brain. 2016;139(Pt 2):481-494. DOI: 10.1093/brain/awv346
  110. 110. Tofaris GK. A critical assessment of exosomes in the pathogenesis and stratification of Parkinson’s disease. Journal of Parkinson’s Disease. 2017;7:569-576. DOI: 10.3233/JPD-171176
  111. 111. Gui Y, Liu H, Zhang L, Lv W, Hu X. Altered microRNA profiles in cerebrospinal fluid exosome in Parkinson disease and Alzheimer disease. Oncotarget. 2015;6:37043-37053. DOI: 10.18632/oncotarget.6158
  112. 112. Fraser KB, Moehle MS, Daher JPL, Webber PJ, Williams JY, Stewart CA, et al. LRRK2 secretion in exosomes is regulated by 14-3-3. Human Molecular Genetics. 2013;22:4988-5000. DOI: 10.1093/hmg/ddt346
  113. 113. Walker LC. Prion-like mechanisms in Alzheimer disease. Handbook of Clinical Neurology. 2018;153:303-319. DOI: 10.1016/B978-0-444-63945-5.00016-7
  114. 114. Jan AT, Malik MA, Rahman S, Yeo HR, Lee EJ, Abdullah TS, et al. Perspective insights of exosomes in neurodegenerative diseases: A critical appraisal. Frontiers in Aging Neuroscience. 2017;9:317. DOI: 10.3389/fnagi.2017.00317
  115. 115. Beach TG. A review of biomarkers for neurodegenerative disease: Will they swing us across the valley? Neurology and Therapy. 2017;6(Suppl 1):5-13. DOI: 10.1007/s40120-017-0072-x
  116. 116. Lin J, Li J, Huang B, Liu J, Chen X, Chen X-M, et al. Exosomes: Novel biomarkers for clinical diagnosis. ScientificWorldJournal. 2015;2015:657086. DOI: 10.1155/2015/657086
  117. 117. Soria FN, Pampliega O, Bourdenx M, Meissner WG, Bezard E, Dehay B. Exosomes, an unmasked culprit in neurodegenerative diseases. Frontiers in Neuroscience. 2017;11:26. DOI: 10.3389/FNINS.2017.00026
  118. 118. Otero-Ortega L, Gómez de Frutos MC, Laso-García F, Rodríguez-Frutos B, Medina-Gutiérrez E, López JA, et al. Exosomes promote restoration after an experimental animal model of intracerebral hemorrhage. Journal of Cerebral Blood Flow and Metabolism. 2018;38:767-779. DOI: 10.1177/0271678X17708917
  119. 119. Doeppner TR, Herz J, Görgens A, Schlechter J, Ludwig A-K, Radtke S, et al. Extracellular vesicles improve post-stroke neuroregeneration and prevent postischemic immunosuppression. Stem Cells Translational Medicine. 2015;4:1131-1143. DOI: 10.5966/sctm.2015-0078
  120. 120. Xin H, Li Y, Liu Z, Wang X, Shang X, Cui Y, et al. MiR-133b promotes neural plasticity and functional recovery after treatment of stroke with multipotent mesenchymal stromal cells in rats via transfer of exosome-enriched extracellular particles. Stem Cells. 2013;31:2737-2746. DOI: 10.1002/stem.1409
  121. 121. Maestrini I, Strbian D, Gautier S, Haapaniemi E, Moulin S, Sairanen T, et al. Higher neutrophil counts before thrombolysis for cerebral ischemia predict worse outcomes. Neurology. 2015;85:1408-1416. DOI: 10.1212/WNL.0000000000002029
  122. 122. Webb RL, Kaiser EE, Jurgielewicz BJ, Spellicy S, Scoville SL, Thompson TA, et al. Human neural stem cell extracellular vesicles improve recovery in a porcine model of ischemic stroke. Stroke. 2018;49:1248-1256. DOI: 10.1161/STROKEAHA.117.020353
  123. 123. Li Y, Cheng Q , Hu G, Deng T, Wang Q , Zhou J, et al. Extracellular vesicles in mesenchymal stromal cells: A novel therapeutic strategy for stroke. Experimental and Therapeutic Medicine. 2018;15:4067-4079. DOI: 10.3892/etm.2018.5993
  124. 124. Lin S-S, Zhu B, Guo Z-K, Huang G-Z, Wang Z, Chen J, et al. Bone marrow mesenchymal stem cell-derived microvesicles protect rat pheochromocytoma PC12 cells from glutamate-induced injury via a PI3K/Akt dependent pathway. Neurochemical Research. 2014;39:922-931. DOI: 10.1007/s11064-014-1288-0
  125. 125. Xin H, Li Y, Buller B, Katakowski M, Zhang Y, Wang X, et al. Exosome-mediated transfer of miR-133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth. Stem Cells. 2012;30:1556-1564. DOI: 10.1002/stem.1129
  126. 126. Liu J, Kuwabara A, Kamio Y, Hu S, Park J, Hashimoto T, et al. Human mesenchymal stem cell-derived microvesicles prevent the rupture of intracranial aneurysm in part by suppression of mast cell activation via a PGE2-dependent mechanism. Stem Cells. 2016;34:2943-2955. DOI: 10.1002/stem.2448

Written By

Laura Cristina Ceafalan, Octavian Costin Ioghen, Daciana Silvia Marta, Alina Constantin, Nicoleta Alexandru, Miruna Nemecz, Gabriela Tanko, Alexandru Filippi, Stefania Lucia Magda, Florina Bojin, Virgil Paunescu, Dragos Vinereanu, Adriana Georgescu and Mihaela Gherghiceanu

Submitted: 10 April 2019 Reviewed: 30 April 2019 Published: 05 July 2019