Next Article in Journal
Vaporized Hydrogen Peroxide and Ozone Gas Synergistically Reduce Prion Infectivity on Stainless Steel Wire
Next Article in Special Issue
Exosomes: Small EVs with Large Immunomodulatory Effect in Glioblastoma
Previous Article in Journal
An Arabidopsis Oxalyl-CoA Decarboxylase, AtOXC, Is Important for Oxalate Catabolism in Plants
Previous Article in Special Issue
Proteomics Profiling of Neuron-Derived Small Extracellular Vesicles from Human Plasma: Enabling Single-Subject Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Secreted Extracellular Vesicle Molecular Cargo as a Novel Liquid Biopsy Diagnostics of Central Nervous System Diseases

1
Cancer Biology & Epigenetics Group, Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
2
Biobank, Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
3
Disease Networks Research Unit, Laboratory of Developmental Biology, Faculty of Biochemistry and Molecular Medicine, Infotech Oulu, Kvantum Institute, University of Oulu, 90570 Oulu, Finland
4
Psychology Service, Portuguese Oncology Institute of Porto (IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
5
BioMark Sensor Research/Centre of Biological Engineering of Minho University, Campus de Gualtar, 4710-057 Braga, Portugal
6
Department of Chemical Engineering, Faculty of Sciences and Technology, University of Coimbra, Rua Sílvio Lima, 3030-790 Coimbra, Portugal
7
Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(6), 3267; https://doi.org/10.3390/ijms22063267
Submission received: 17 February 2021 / Revised: 18 March 2021 / Accepted: 20 March 2021 / Published: 23 March 2021

Abstract

:
Secreted extracellular vesicles (EVs) are heterogeneous cell-derived membranous granules which carry a large diversity of molecules and participate in intercellular communication by transferring these molecules to target cells by endocytosis. In the last decade, EVs’ role in several pathological conditions, from etiology to disease progression or therapy evasion, has been consolidated, including in central nervous system (CNS)-related disorders. For this review, we performed a systematic search of original works published, reporting the presence of molecular components expressed in the CNS via EVs, which have been purified from plasma, serum or cerebrospinal fluid. Our aim is to provide a list of molecular EV components that have been identified from both nonpathological conditions and the most common CNS-related disorders. We discuss the methods used to isolate and enrich EVs from specific CNS-cells and the relevance of its components in each disease context.

1. Introduction

Extracellular vesicles (EVs) are cell-derived, membrane-bound structures, secreted into the extracellular environment by most, if not all, cell types. There is mounting evidence that EVs take part in control of normal physiological processes, in homeostasis control and in cell-to-cell communication within tissues and organs. Based on development of the EV field, different types of vesicles have been described, and include apoptotic bodies which range in size from 50 to 5000 nm, microvesicles (50–1000 nm), exosomes (40–200 nm) and the more recently recognized exomeres [1,2,3,4].
The EVs have started to attract attention also as potential disease biomarkers and as new ways to study the control of cell biology. They seem to offer diagnostic capacity of organ dysfunctions via the body fluids as the components of the “liquid biopsy” type of sample. For example, solid tumors exhibit the elusive nature of transformed cells. They grow into organs but can metastasize to distant locations. The original tumor cells and their metastases secrete EVs and this has offered a promising way to diagnose cancers from its inception [5]. By studying the detailed molecular content of the cancer cell secreted EVs, tumor specific signatures may be defined and information about the changes in the associated biological processes gained.
In central nervous system (CNS) diseases, such as Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, amyotrophic lateral sclerosis, gliomas, traumatic brain injury, stroke, schizophrenia and major depressive disorders, neuroscientists and neurologists cannot easily access the diseased tissues for diagnostic purposes. If brain biopsy is to be taken, it might not be representative of the common neurological disorder. Therefore, if the EVs that would be released from the CNS to systemic circulation could be identified reliably, this would provide a breakthrough to advance diagnostics of neurological diseases [6].
One of the challenges when working with EVs, particularly for CNS and CNS-related pathological conditions, is to ensure that the CNS-derived EVs can be isolated from the biological fluids (e.g., plasma (containing blood clotting agents), serum (without blood clotting agents) or cerebrospinal fluid (CSF)) and that these indeed would have originated from neural cells. Since EVs also contain the HLA-associated, self-depicting peptides and wealth of other types of cargo proteins of donor cells, such EV molecules may serve to identify specific organ level signatures, thus portraying the cell of origin. Since the EVs also have the ability to cross the blood–brain barrier (BBB) this raises the possibility that EV molecular content in biological fluids may reflect certain neuronal processes, as well [7]. Currently a relatively large body of literature documents the importance of EVs, but none has yet systematically summarized EVs molecular content in association with CNS and common CNS-related diseases. In this review, we provide a catalogue of CNS-derived EVs’ molecular cargo, both in nonpathological and selected pathological conditions.

2. Methods

A PubMed search was conducted, with the query (extracellular vesicles OR exosomes OR microvesicles) AND (markers OR biomarkers) AND (brain OR central nervous system), with no time interval restraints. Only original studies written in English were considered. The retrieved records from the search were collected into the reference manager Endnote. All abstracts were critically assessed to select only those providing meaningful information related to the topic. Only studies using human cerebrospinal fluid (CSF), serum or plasma samples were included. A flow diagram with a summary of the methodology is provided in Figure 1.

3. Methods and Factors for Isolation of Central Nervous System Extracellular Vesicles in Liquid Biopsies

Currently, as EVs are known to be able to cross the BBB, they are considered as potential biomarkers for monitoring CNS disorders as well as specific treatment responses. Of particular interest is the ability to characterize EVs based on their source, which may provide insight into the target disease. Although the majority of EVs research is performed through cell culture, the number of reports aiming at identifying biological fluids’ EVs originated in different cell types is rising. Multiple methods (including ultracentrifugation, immunomagnetic beads, and size exclusion chromatography) can be used to separate mixed EVs (from multiple cell types) from different biological fluids [8]. This step may be taken further by using cell-specific protein markers to enrich for EVs of specific cell types from a mixed population of vesicles.
Specifically, EVs from the CNS are secreted from almost all cell types, including various types of neurons, astrocytes, oligodendrocytes, microglia and Schwann cells, and endothelial cells. EVs carry a variety of molecules related to neuronal function and neurotransmission in the brain. This contributes to the communication between nerve cells (for example, neuron–glia interaction), synaptic plasticity, and neuron development. Interestingly, Fiandaca et al. developed an immunoprecipitation-based method to isolate EVs rich in neuron sources from blood. This technology has been also used by others and consists of two steps: a preliminary separation of total EVs from plasma or serum samples using commercially available polymers for high-throughput particle precipitation, and immunoprecipitation with biotinylated antibodies against the neuron surface marker neuronal markers L1 cell adhesion molecule (L1CAM, CD171) [9]. Furthermore, L1CAM and the GluR2/3 subunits of glutamate receptors have been used for the identification of EVs that may be released from developing and mature hippocampal neurons [10,11]. Hence, using this method, EVs of neuronal origin may be enriched and evaluated based on proteins, lipids and nucleic acids.
Although neuron-derived EVs are a common target in several studies, researchers are also interested in isolating EVs originating from other CNS cells, aiming also to translate via the EVs the origin of CNS-related diseases. The above-mentioned methodology can also be used to enrich for EVs from other cellular origins with antibodies against specific cell surface components. For example, for astrocytic-derived EVs enrichment, glial fibrillary acidic protein (GFAP), glutamine aspartate transporter (GLAST), and glutamine synthetase (GLUL) have been suggested to be specific enough to obtain CNS related EVs [12]. Additionally, for EVs originated in oligodendrocytes, myelin proteolipid protein (PLP) and 2′,3′-cyclic nucleotide 3′-phosphodiesterase (CNP) have been proposed as specific enough to identify the cell from which the EVs derived [13] (Figure 2).
The application of specific immunoaffinity-based methodology for the isolation of CNS-related EVs in biological fluids, applied to different CNS diseases research, has led to promising results [9,14]. Once the enriched fraction of EVs is obtained, either surface or cargo content can be analyzed using different approaches according to the molecular target of interest, such as proteomics (e.g., mass spectrometry, Western blot or enzyme-linked immunosorbent assays) and/or transcriptomics methodologies (e.g., high-throughput RNA sequencing or RT-qPCR). This opens a new avenue for research, using CNS-derived EVs as vehicles for disease monitoring.

4. Central Nervous System Disease-Related Biomarkers in Extracellular Vesicles

CNS-related disorders, including neurodegenerative diseases, such as Alzheimer’s (AD) and Parkinson’s (PD) diseases, malignant tumors, such as gliomas, and psychiatric disorders, are very different pathological conditions, but share common features: anatomical origin and difficult early diagnosis. The existing liquid biopsies and imaging biomarkers are rather imperfect, because their dynamic range does not cover the entire course of the disease, and their classification accuracy is lower than the level accepted for clinical practice, thus, precluding their routine implementation. Thus, the use of CNS-enriched EVs and their molecular content as biomarkers for these specific pathological conditions may revolutionize the clinical management of these patients. Indeed, a growing number of studies within this field have been published in the last few years. To simplify, a summary of the main publications retrieved from the database query can be found in Table 1 and Table 2, referring to either cargo or surface molecules, respectively [9,12,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97] (Figure 3).
Alzheimer’s disease (AD) is neurodegenerative disorder characterized by memory loss and behavioral changes. EVs have been studied as a potential diagnostic marker for AD. This disease has a complex progression including early development of neuronal dysplasia, angiogenic changes, release of inflammatory mediators by CNS glial and peripheral immune cells, and development of extracellular amyloid-β (Aβ) fibrils that deposit in the brain as amyloid plaques, and impair synaptic plasticity [98]. At the molecular level, the extracellular deposition of insoluble amyloid-β Aβ peptide plaques (39–43 amino acids produced by amyloid precursor protein, APP, peptides) occurs, which can interact with regulatory proteins to phosphorylate the microtubule-associated protein tau. The increase of phosphorylated tau protein (P-S396-tau, P-T181-tau) and Aβ42 detected in plasma EVs is associated with increased risk for AD [98]. Of note, both Aβ and tau are secreted by neurons through the release of EVs [12,26,36,56,90]. Moreover, some authors also studied molecules found in EVs from patients suffering from mild cognitive impairment. Their memory has deficits that do not significantly affect daily functioning, but are often regarded and treated as early phase AD [52,55,99].
Parkinson’s disease (PD) is other common neurodegenerative disorder. Although PD can be described more correctly as a syndrome caused by different genetic and epigenetic alterations, a shared downstream result is the degeneration of dopamine-releasing axon terminals in the striatum and of corresponding neurons in the substantia nigra, which leads to impairment of motor and speech skills [100]. EVs have also been used as a diagnostic platform for PD. Indeed, increased α-synuclein levels in plasma EVs were associated with PD, and disease clinical severity, although results are inconsistent [18,72,88]. Moreover, reduced levels of CSF’s EVs containing apolipoprotein A1 were associated with a higher risk of PD [21].
Multiple sclerosis (MS) and amyotrophic lateral sclerosis (ALS) are very different demyelinating diseases. MS is an autoimmune disease that affects the myelin sheath, which insulates nerve cell fibers in the brain and spinal cord [101], whereas ALS is a motor neuron disease that mainly affects the brain and spinal cord motor neuron cells [102]. For both diseases, several efforts have been made to find disease-related biomarkers in EVs isolated from patients’ biological fluids, with a special emphasis in non-coding RNAs.
Non-coding RNAs (ncRNAs) are also generally found in circulating EVs. There are several ncRNA categories, commonly classified according to their size: the long ncRNAs (lncRNAs) with more than 200 nt, and the small ncRNAs (sncRNAs), including microRNAs (miRs), which present less than 200 nt [103,104]. These ncRNAs have been studied as specific disease “signatures” found in EVs in various disorders including MS/ALS. In fact, for all major group of CNS-related diseases mentioned in this review, published data on EVs ncRNAs have been assessed. For instance, in a recent study by Ebrahimkhani S. et al., the utility of serum exosome miRs as disease biomarkers of MS patients under treatment with fingolimod was assessed, and they found that several combinations of two or three miRNAs could discriminate active from quiescent disease with more than 90% accuracy [79]. Likewise, for ALS, Banack et al. found that eight miRNAs, isolated from neural-enriched EVs, significantly distinguished ALS patients from controls, and thus might assist in early diagnosis of this disease [75].
Gliomas are glial cell-derived brain tumors, classified according to cell type, including ependymomas (ependymal cells), oligodendrogliomas (oligodendrocytes) and astrocytomas (astrocytes). The most common and malignant primary brain tumor, the grade IV astrocytoma/glioblastoma multiform belongs to this group of tumors [105]. Glioblastoma-specific or glioblastoma-rich protein and genetic material can be detected in EVs isolated from the biological fluids of glioblastoma’s patients. The usefulness of using EVs as platforms for the analysis of specific biomarkers, which can help predict the disease outcome and prognosis of GM patients, have been recently established. For example, EGFRvIII is the oncogenic form of epidermal growth factor receptor (EGFR), never found in normal tissues. According to various reports, EGFRvIII protein, mRNA and DNA have been detected in plasma EVs isolated from patients with glioblastoma carrying EGFRvIII, but not in EVs isolated from noncancer patients [28,41,44]. Similarly, multiple studies have also shown that miR-21 levels are higher in EVs isolated from the serum and CSF of patients with glioblastoma. This finding is supported by the higher levels of miR-21 also described in clinical glioblastoma specimens [20,27,46].
Traumatic brain injury (TBI), also known as intracranial injury, is a brain damage caused by external forces [106]. TBI classification can be based on severity (from mild to severe TBI), mechanism (closed or penetrating head injury), or other characteristics (for example, occurring in a specific location or widespread area). TBI usually causes neurological sequels, which can be seen even in mild TBI. Mild TBI may lead to acute symptoms, including chronic traumatic encephalopathy, cognitive impairment, dementia, movement disorders, and motor neuron dysfunction [107]. One of the main clinical challenges in TBI is to accurately identify its occurrence, and to determine the extent of CNS damage. Peltz et al. showed that in war veterans with a history of TBI, CNS-enriched exosome concentration of pTAU, NfL, IL-6, TNFa are associated with cognitive impairment, which may assist clinicians in choosing a more appropriate treatment schedule and follow-up [89].
Psychiatric disorders, like schizophrenia (SCZ) or chronic depression, are estimated to affect a major percentage of the world population and are very difficult to diagnose and manage. Schizophrenia is a neurological disorder characterized by behavioral deficits, associated with impaired locomotor activity and cognitive defects. The diagnosis of schizophrenia is associated with demonstrable alterations in brain structure and changes in dopamine and glutamate neurotransmission in the cortex [108]. In this field, there is a demand for the discovery of biomarkers, which, if successful, can support clinicians in personalized treatment strategies. In this context, Du Y. et al. showed that the expression levels of specific miRs in EVs isolated from the blood of SCZ patients were sensitive to long-term medication [58].
Stroke, which can be categorized into ischemic or hemorrhagic stroke, affects 13.7 million people worldwide each year, and is the second leading cause of death, with 5.5 million deaths each year [109]. A stroke occurs when the blood supply to part of the brain is interrupted or reduced, limiting brain tissue from obtaining oxygen and nutrients, and generally causing brain damage [110]. Several authors explored the importance of the innate immune response as a contributor to the inflammatory response after stroke [111,112,113]. In this regard, Kerr et al. used a technique designated Simple Plex assay (an immunoassay in a microfluidic cartridge) to determine the presence and concentration of some inflammasome proteins in serum EVs of stroke patients [38]. The authors found that protein levels of ASC (caspase-recruitment domain) remained higher in serum-derived EVs from stroke samples, when compared to controls, and performed well as a potential biomarker for this pathological condition [38].
Major depressive disorder (MDD) is a debilitating mental disease characterized by persistent low mood. It affects the behavior as well as various physical functions, such as appetite and sleep [114]. Although the understanding of the neurobiology of MDD has improved in recent years, the knowledge of the mechanisms that may explain the most relevant aspects of the disease is limited. Nonetheless, MDD was associated with smaller hippocampal volumes, changes in activation or connectivity of neural networks, and also changes in the main neurobiological systems that mediate the stress response, including the hypothalamic–pituitary–adrenal (HPA) axis, autonomic nervous system, and immune system [115,116]. The identification of susceptibility biomarkers that might identify individuals prone to depression and disease recurrence would improve treatment and allow for recurrence prevention. A series of studies by different research teams have implicated a systemic metabolic dysfunction known as insulin resistance in the pathophysiology and treatment of disorders of mood and cognition, including MDD [117,118]. Therefore, the insulin-receptor (IR) investigated in these studies was identified in brain areas associated with mood and cognition [119]. Indeed, Nasca et al. recently found that insulin-receptor substrate-1 (IRS-1) was highly abundant in neuronal-derived EVs isolated from plasma samples of MDD patients, compared to nonpathological controls, and that higher IRS-1 levels were associated with suicidality and anhedonia in those individuals. This data could lead to an improved strategy in the treatment of MDD patients [86].

5. Future Perspectives and Conclusions

Research based on EVs, specifically neuronal-derived EVs, is rapidly moving forward. We have now recognized a wide range of biological processes mediated by these molecules, and its importance into cell–cell communication, as well as disease spread within the CNS. With increasing knowledge about EVs from specific neurological diseases and the advances in technologies used to analyze these nanostructures, research is now moving towards clinical translation for biomarker platforms. For this, established and validated biomarkers are needed. Indeed, various single reports based on small cohorts have been published, but very few were validated by larger, cross-sectional investigations. The promising results so far point towards a future where brain-derived EVs could be used for not only for diagnosis of CNS-related disorders, but also for patient monitoring, and for studying the influence of the release of this specific EVs in behavioral and mental health related conditions.
Overall, EVs produced in the brain and circulating in peripheral fluids have become an incredible window to the brain, which is expected to assess the disease status of a given patient with great accuracy and simplicity. In the near future, one can anticipate that a relevant biomarker platform may move quickly into clinical context, to provide an enormous amount of information that may assist in clinical patients’ follow-up. This may occur at the physician’s office, as many technological solutions exist today to monitor such biomarkers using point-of-care devices. In a long-term perspective, one may anticipate that the information deriving from EVs may also act as an early disease warning system. It is only natural that the biological/biochemical information of such circulating EVs shall be better and more significant in time, considering the huge developments expected in the fields of nanotechnology, bioengineering and artificial intelligence. Specifically, when these are combined for the same purpose, even lower concentrations of a given pool of biomarkers in very complex samples might be detected. Finally, it will be indeed a breakthrough when such advanced biomarker panel from circulating EVs (that may well derive from CNS) becomes a tool to prevent disease progression.

Author Contributions

Conceptualization, S.M.-R., R.H. and C.J.; Methodology, S.M.-R., C.C.-M. and J.P.; Data collection, S.M.-R.; Writing—Original draft Preparation and Editing, S.M.-R.; Writing—Reviewing and Editing, G.B., S.J.V., E.R.S., G.S., R.H. and C.J.; Supervision, R.H. and C.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the MindGaP-H2020-FETOPEN-2018-2020, Grant agreement ID: 829040. S.M.-R., C.C.-M., and J.P. hold a fellowship from MindGaP.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Samanta, S.; Rajasingh, S.; Drosos, N.; Zhou, Z.; Dawn, B.; Rajasingh, J. Exosomes: New molecular targets of diseases. Acta Pharmacol. Sin. 2018, 39, 501–513. [Google Scholar] [CrossRef]
  2. Lee, Y.; El Andaloussi, S.; Wood, M.J. Exosomes and microvesicles: Extracellular vesicles for genetic information transfer and gene therapy. Hum. Mol. Genet. 2012, 21, R125–R134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Gheinani, A.H.; Vogeli, M.; Baumgartner, U.; Vassella, E.; Draeger, A.; Burkhard, F.C.; Monastyrskaya, K. Improved isolation strategies to increase the yield and purity of human urinary exosomes for biomarker discovery. Sci. Rep. 2018, 8, 3945. [Google Scholar] [CrossRef]
  4. Zhang, H.; Lyden, D. Asymmetric-flow field-flow fractionation technology for exomere and small extracellular vesicle separation and characterization. Nat. Protoc. 2019, 14, 1027–1053. [Google Scholar] [CrossRef]
  5. Dos Anjos Pultz, B.; Andres Cordero da Luz, F.; Socorro Faria, S.; Peixoto Ferreira de Souza, L.; Cristina Brigido Tavares, P.; Alonso Goulart, V.; Fontes, W.; Ricardo Goulart, L.; Jose Barbosa Silva, M. The multifaceted role of extracellular vesicles in metastasis: Priming the soil for seeding. Int. J. Cancer 2017, 140, 2397–2407. [Google Scholar] [CrossRef] [Green Version]
  6. Shi, M.; Sheng, L.; Stewart, T.; Zabetian, C.P.; Zhang, J. New windows into the brain: Central nervous system-derived extracellular vesicles in blood. Prog. Neurobiol. 2019, 175, 96–106. [Google Scholar] [CrossRef] [PubMed]
  7. Matsumoto, J.; Stewart, T.; Banks, W.A.; Zhang, J. The Transport Mechanism of Extracellular Vesicles at the Blood-Brain Barrier. Curr. Pharm. Des. 2017, 23, 6206–6214. [Google Scholar] [CrossRef]
  8. Brennan, K.; Martin, K.; FitzGerald, S.P.; O’Sullivan, J.; Wu, Y.; Blanco, A.; Richardson, C.; Mc Gee, M.M. A comparison of methods for the isolation and separation of extracellular vesicles from protein and lipid particles in human serum. Sci. Rep. 2020, 10, 1039. [Google Scholar] [CrossRef] [Green Version]
  9. Fiandaca, M.S.; Kapogiannis, D.; Mapstone, M.; Boxer, A.; Eitan, E.; Schwartz, J.B.; Abner, E.L.; Petersen, R.C.; Federoff, H.J.; Miller, B.L.; et al. Identification of preclinical Alzheimer’s disease by a profile of pathogenic proteins in neurally derived blood exosomes: A case-control study. Alzheimer’s Dement. 2015, 11, 600–607e601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Lachenal, G.; Pernet-Gallay, K.; Chivet, M.; Hemming, F.J.; Belly, A.; Bodon, G.; Blot, B.; Haase, G.; Goldberg, Y.; Sadoul, R. Release of exosomes from differentiated neurons and its regulation by synaptic glutamatergic activity. Mol. Cell Neurosci. 2011, 46, 409–418. [Google Scholar] [CrossRef] [Green Version]
  11. Faure, J.; Lachenal, G.; Court, M.; Hirrlinger, J.; Chatellard-Causse, C.; Blot, B.; Grange, J.; Schoehn, G.; Goldberg, Y.; Boyer, V.; et al. Exosomes are released by cultured cortical neurones. Mol. Cell Neurosci. 2006, 31, 642–648. [Google Scholar] [CrossRef] [PubMed]
  12. Goetzl, E.J.; Mustapic, M.; Kapogiannis, D.; Eitan, E.; Lobach, I.V.; Goetzl, L.; Schwartz, J.B.; Miller, B.L. Cargo proteins of plasma astrocyte-derived exosomes in Alzheimer’s disease. FASEB J. 2016, 30, 3853–3859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kramer-Albers, E.M.; Bretz, N.; Tenzer, S.; Winterstein, C.; Mobius, W.; Berger, H.; Nave, K.A.; Schild, H.; Trotter, J. Oligodendrocytes secrete exosomes containing major myelin and stress-protective proteins: Trophic support for axons? Proteomics Clin. Appl. 2007, 1, 1446–1461. [Google Scholar] [CrossRef]
  14. Mustapic, M.; Eitan, E.; Werner, J.K., Jr.; Berkowitz, S.T.; Lazaropoulos, M.P.; Tran, J.; Goetzl, E.J.; Kapogiannis, D. Plasma extracellular vesicles enriched for neuronal origin: A potential window into brain pathologic processes. Front. Neurosci. 2017, 11, 278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Mobarrez, F.; Nybom, R.; Johansson, V.; Hultman, C.M.; Wallén, H.; Landén, M.; Wetterberg, L. Microparticles and microscopic structures in three fractions of fresh cerebrospinal fluid in schizophrenia: Case report of twins. Schizophr. Res. 2013, 143, 192–197. [Google Scholar] [CrossRef] [PubMed]
  16. Kanhai, D.A.; de Kleijn, D.P.; Kappelle, L.J.; Uiterwaal, C.S.; van der Graaf, Y.; Pasterkamp, G.; Geerlings, M.I.; Visseren, F.L. 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. [Google Scholar] [CrossRef] [Green Version]
  17. Manterola, L.; Guruceaga, E.; Gállego Pérez-Larraya, J.; González-Huarriz, M.; Jauregui, P.; Tejada, S.; Diez-Valle, R.; Segura, V.; Samprón, N.; Barrena, C.; et al. A small noncoding RNA signature found in exosomes of GBM patient serum as a diagnostic tool. Neuro. Oncol. 2014, 16, 520–527. [Google Scholar] [CrossRef]
  18. Shi, M.; Liu, C.; Cook, T.J.; Bullock, K.M.; Zhao, Y.; Ginghina, C.; Li, Y.; Aro, P.; Dator, R.; He, C.; et al. Plasma exosomal α-synuclein is likely CNS-derived and increased in Parkinson’s disease. Acta Neuropathol. 2014, 128, 639–650. [Google Scholar] [CrossRef]
  19. Shao, H.; Chung, J.; Lee, K.; Balaj, L.; Min, C.; Carter, B.S.; Hochberg, F.H.; Breakefield, X.O.; Lee, H.; Weissleder, R. Chip-based analysis of exosomal mRNA mediating drug resistance in glioblastoma. Nat. Commun. 2015, 6, 6999. [Google Scholar] [CrossRef] [Green Version]
  20. Shi, R.; Wang, P.Y.; Li, X.Y.; Chen, J.X.; Li, Y.; Zhang, X.Z.; Zhang, C.G.; Jiang, T.; Li, W.B.; Ding, W.; et al. Exosomal levels of miRNA-21 from cerebrospinal fluids associated with poor prognosis and tumor recurrence of glioma patients. Oncotarget 2015, 6, 26971–26981. [Google Scholar] [CrossRef] [Green Version]
  21. Yang, Y.; Keene, C.D.; Peskind, E.R.; Galasko, D.R.; Hu, S.C.; Cudaback, E.; Wilson, A.M.; Li, G.; Yu, C.E.; Montine, K.S.; et al. Cerebrospinal fluid particles in Alzheimer disease and Parkinson disease. J. Neuropathol. Exp. Neurol. 2015, 74, 672–687. [Google Scholar] [CrossRef] [Green Version]
  22. Goetzl, E.J.; Kapogiannis, D.; Schwartz, J.B.; Lobach, I.V.; Goetzl, L.; Abner, E.L.; Jicha, G.A.; Karydas, A.M.; Boxer, A.; Miller, B.L. Decreased synaptic proteins in neuronal exosomes of frontotemporal dementia and Alzheimer’s disease. FASEB J. 2016, 30, 4141–4148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Ji, Q.; Ji, Y.; Peng, J.; Zhou, X.; Chen, X.; Zhao, H.; Xu, T.; Chen, L.; Xu, Y. Increased brain-specific MiR-9 and MiR-124 in the serum exosomes of acute ischemic stroke patients. PLoS ONE 2016, 11, e0163645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Lee, J.; McKinney, K.Q.; Pavlopoulos, A.J.; Han, M.H.; Kim, S.H.; Kim, H.J.; Hwang, S. Exosomal proteome analysis of cerebrospinal fluid detects biosignatures of neuromyelitis optica and multiple sclerosis. Clin. Chim. Acta 2016, 462, 118–126. [Google Scholar] [CrossRef]
  25. Shi, M.; Kovac, A.; Korff, A.; Cook, T.J.; Ginghina, C.; Bullock, K.M.; Yang, L.; Stewart, T.; Zheng, D.; Aro, P.; et al. CNS tau efflux via exosomes is likely increased in Parkinson’s disease but not in Alzheimer’s disease. Alzheimer’s Dement. 2016, 12, 1125–1131. [Google Scholar] [CrossRef] [Green Version]
  26. Winston, C.N.; Goetzl, E.J.; Akers, J.C.; Carter, B.S.; Rockenstein, E.M.; Galasko, D.; Masliah, E.; Rissman, R.A. Prediction of conversion from mild cognitive impairment to dementia with neuronally derived blood exosome protein profile. Alzheimer’s Dement. 2016, 3, 63–72. [Google Scholar] [CrossRef] [Green Version]
  27. Akers, J.C.; Hua, W.; Li, H.; Ramakrishnan, V.; Yang, Z.; Quan, K.; Zhu, W.; Li, J.; Figueroa, J.; Hirshman, B.R.; et al. A cerebrospinal fluid microRNA signature as biomarker for glioblastoma. Oncotarget 2017, 8, 68769–68779. [Google Scholar] [CrossRef] [PubMed]
  28. Figueroa, J.M.; Skog, J.; Akers, J.; Li, H.; Komotar, R.; Jensen, R.; Ringel, F.; Yang, I.; Kalkanis, S.; Thompson, R.; et al. Detection of wild-type EGFR amplification and EGFRvIII mutation in CSF-derived extracellular vesicles of glioblastoma patients. Neuro. Oncol. 2017, 19, 1494–1502. [Google Scholar] [CrossRef] [PubMed]
  29. Mullins, R.J.; Mustapic, M.; Goetzl, E.J.; Kapogiannis, D. Exosomal biomarkers of brain insulin resistance associated with regional atrophy in Alzheimer’s disease. Hum. Brain Mapp. 2017, 38, 1933–1940. [Google Scholar] [CrossRef] [Green Version]
  30. Wang, S.; Liu, Z.; Ye, T.; Mabrouk, O.S.; Maltbie, T.; Aasly, J.; West, A.B. Elevated LRRK2 autophosphorylation in brain-derived and peripheral exosomes in LRRK2 mutation carriers. Acta Neuropathol. Commun. 2017, 5, 86. [Google Scholar] [CrossRef]
  31. Welton, J.L.; Loveless, S.; Stone, T.; von Ruhland, C.; Robertson, N.P.; Clayton, A. Cerebrospinal fluid extracellular vesicle enrichment for protein biomarker discovery in neurological disease; multiple sclerosis. J. Extracell. Vesicles 2017, 6, 1369805. [Google Scholar] [CrossRef] [PubMed]
  32. Yan, S.; Zhang, H.; Xie, W.; Meng, F.; Zhang, K.; Jiang, Y.; Zhang, X.; Zhang, J. Altered microRNA profiles in plasma exosomes from mesial temporal lobe epilepsy with hippocampal sclerosis. Oncotarget 2017, 8, 4136–4146. [Google Scholar] [CrossRef]
  33. Drusco, A.; Fadda, P.; Nigita, G.; Fassan, M.; Bottoni, A.; Gardiman, M.P.; Sacchi, D.; Calore, F.; Carosi, M.; Antenucci, A.; et al. circulating micrornas predict survival of patients with tumors of glial origin. EBioMedicine 2018, 30, 105–112. [Google Scholar] [CrossRef]
  34. Galazka, G.; Mycko, M.P.; Selmaj, I.; Raine, C.S.; Selmaj, K.W. Multiple sclerosis: Serum-derived exosomes express myelin proteins. Mult. Scler. 2018, 24, 449–458. [Google Scholar] [CrossRef]
  35. Goetzl, E.J.; Abner, E.L.; Jicha, G.A.; Kapogiannis, D.; Schwartz, J.B. Declining levels of functionally specialized synaptic proteins in plasma neuronal exosomes with progression of Alzheimer’s disease. FASEB J. 2018, 32, 888–893. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Guix, F.X.; Corbett, G.T.; Cha, D.J.; Mustapic, M.; Liu, W.; Mengel, D.; Chen, Z.; Aikawa, E.; Young-Pearse, T.; Kapogiannis, D.; et al. Detection of aggregation-competent tau in neuron-derived extracellular vesicles. Int. J. Mol. Sci. 2018, 19, 663. [Google Scholar] [CrossRef] [Green Version]
  37. Huang, K.; Fang, C.; Yi, K.; Liu, X.; Qi, H.; Tan, Y.; Zhou, J.; Li, Y.; Liu, M.; Zhang, Y.; et al. The role of PTRF/Cavin1 as a biomarker in both glioma and serum exosomes. Theranostics 2018, 8, 1540–1557. [Google Scholar] [CrossRef]
  38. Kerr, N.; García-Contreras, M.; Abbassi, S.; Mejias, N.H.; Desousa, B.R.; Ricordi, C.; Dietrich, W.D.; Keane, R.W.; de Rivero Vaccari, J.P. inflammasome proteins in serum and serum-derived extracellular vesicles as biomarkers of stroke. Front. Mol. Neurosci. 2018, 11, 309. [Google Scholar] [CrossRef] [Green Version]
  39. Kuwano, N.; Kato, T.A.; Mitsuhashi, M.; Sato-Kasai, M.; Shimokawa, N.; Hayakawa, K.; Ohgidani, M.; Sagata, N.; Kubo, H.; Sakurai, T.; et al. Neuron-related blood inflammatory markers as an objective evaluation tool for major depressive disorder: An exploratory pilot case-control study. J. Affect. Disord. 2018, 240, 88–98. [Google Scholar] [CrossRef] [PubMed]
  40. Lan, F.; Qing, Q.; Pan, Q.; Hu, M.; Yu, H.; Yue, X. Serum exosomal miR-301a as a potential diagnostic and prognostic biomarker for human glioma. Cell Oncol. 2018, 41, 25–33. [Google Scholar] [CrossRef] [PubMed]
  41. Manda, S.V.; Kataria, Y.; Tatireddy, B.R.; Ramakrishnan, B.; Ratnam, B.G.; Lath, R.; Ranjan, A.; Ray, A. Exosomes as a biomarker platform for detecting epidermal growth factor receptor-positive high-grade gliomas. J. Neurosurg. 2018, 128, 1091–1101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. McKeever, P.M.; Schneider, R.; Taghdiri, F.; Weichert, A.; Multani, N.; Brown, R.A.; Boxer, A.L.; Karydas, A.; Miller, B.; Robertson, J.; et al. MicroRNA expression levels are altered in the cerebrospinal fluid of patients with young-onset Alzheimer’s disease. Mol. Neurobiol. 2018, 55, 8826–8841. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Pieragostino, D.; Cicalini, I.; Lanuti, P.; Ercolino, E.; di Ioia, M.; Zucchelli, M.; Zappacosta, R.; Miscia, S.; Marchisio, M.; Sacchetta, P.; et al. Enhanced release of acid sphingomyelinase-enriched exosomes generates a lipidomics signature in CSF of Multiple Sclerosis patients. Sci. Rep. 2018, 8, 3071. [Google Scholar] [CrossRef] [Green Version]
  44. Reátegui, E.; van der Vos, K.E.; Lai, C.P.; Zeinali, M.; Atai, N.A.; Aldikacti, B.; Floyd, F.P., Jr.; Aimal, H.K.; Thapar, V.; Hochberg, F.H.; et al. Engineered nanointerfaces for microfluidic isolation and molecular profiling of tumor-specific extracellular vesicles. Nat. Commun. 2018, 9, 175. [Google Scholar] [CrossRef] [PubMed]
  45. Ricklefs, F.L.; Alayo, Q.; Krenzlin, H.; Mahmoud, A.B.; Speranza, M.C.; Nakashima, H.; Hayes, J.L.; Lee, K.; Balaj, L.; Passaro, C.; et al. Immune evasion mediated by PD-L1 on glioblastoma-derived extracellular vesicles. Sci. Adv. 2018, 4, eaar2766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Santangelo, A.; Imbrucè, P.; Gardenghi, B.; Belli, L.; Agushi, R.; Tamanini, A.; Munari, S.; Bossi, A.M.; Scambi, I.; Benati, D.; et al. A microRNA signature from serum exosomes of patients with glioma as complementary diagnostic biomarker. J. Neurooncol. 2018, 136, 51–62. [Google Scholar] [CrossRef] [PubMed]
  47. Schneider, R.; McKeever, P.; Kim, T.; Graff, C.; van Swieten, J.C.; Karydas, A.; Boxer, A.; Rosen, H.; Miller, B.L.; Laforce, R., Jr.; et al. Downregulation of exosomal miR-204-5p and miR-632 as a biomarker for FTD: A GENFI study. J. Neurol. Neurosurg. Psychiatr. 2018, 89, 851–858. [Google Scholar] [CrossRef]
  48. Sproviero, D.; La Salvia, S.; Giannini, M.; Crippa, V.; Gagliardi, S.; Bernuzzi, S.; Diamanti, L.; Ceroni, M.; Pansarasa, O.; Poletti, A.; et al. Pathological proteins are transported by extracellular vesicles of sporadic amyotrophic lateral sclerosis patients. Front. Neurosci. 2018, 12, 487. [Google Scholar] [CrossRef]
  49. Tan, S.K.; Pastori, C.; Penas, C.; Komotar, R.J.; Ivan, M.E.; Wahlestedt, C.; Ayad, N.G. Serum long noncoding RNA HOTAIR as a novel diagnostic and prognostic biomarker in glioblastoma multiforme. Mol. Cancer 2018, 17, 74. [Google Scholar] [CrossRef] [Green Version]
  50. Wang, W.; Li, D.B.; Li, R.Y.; Zhou, X.; Yu, D.J.; Lan, X.Y.; Li, J.P.; Liu, J.L. Diagnosis of Hyperacute and Acute Ischaemic Stroke: The potential utility of exosomal MicroRNA-21-5p and MicroRNA-30a-5p. Cerebrovasc. Dis. 2018, 45, 204–212. [Google Scholar] [CrossRef] [Green Version]
  51. Wei, H.; Xu, Y.; Xu, W.; Zhou, Q.; Chen, Q.; Yang, M.; Feng, F.; Liu, Y.; Zhu, X.; Yu, M.; et al. Serum exosomal miR-223 serves as a potential diagnostic and prognostic biomarker for dementia. Neuroscience 2018, 379, 167–176. [Google Scholar] [CrossRef]
  52. Winston, C.N.; Goetzl, E.J.; Baker, L.D.; Vitiello, M.V.; Rissman, R.A. Growth hormone-releasing hormone modulation of neuronal exosome biomarkers in mild cognitive impairment. J. Alzheimer’s Dis. 2018, 66, 971–981. [Google Scholar] [CrossRef]
  53. Agliardi, C.; Guerini, F.R.; Zanzottera, M.; Bianchi, A.; Nemni, R.; Clerici, M. SNAP-25 in serum is carried by exosomes of neuronal origin and is a potential biomarker of Alzheimer’s disease. Mol. Neurobiol. 2019, 56, 5792–5798. [Google Scholar] [CrossRef] [PubMed]
  54. Athauda, D.; Gulyani, S.; Karnati, H.K.; Li, Y.; Tweedie, D.; Mustapic, M.; Chawla, S.; Chowdhury, K.; Skene, S.S.; Greig, N.H.; et al. Utility of neuronal-derived exosomes to examine molecular mechanisms that affect motor function in patients with Parkinson disease: A secondary analysis of the exenatide-PD Trial. JAMA Neurol. 2019, 76, 420–429. [Google Scholar] [CrossRef] [PubMed]
  55. Cha, D.J.; Mengel, D.; Mustapic, M.; Liu, W.; Selkoe, D.J.; Kapogiannis, D.; Galasko, D.; Rissman, R.A.; Bennett, D.A.; Walsh, D.M. miR-212 and miR-132 are downregulated in neurally derived plasma exosomes of Alzheimer’s patients. Front. Neurosci. 2019, 13, 1208. [Google Scholar] [CrossRef] [Green Version]
  56. Cicognola, C.; Brinkmalm, G.; Wahlgren, J.; Portelius, E.; Gobom, J.; Cullen, N.C.; Hansson, O.; Parnetti, L.; Constantinescu, R.; Wildsmith, K.; et al. Novel tau fragments in cerebrospinal fluid: Relation to tangle pathology and cognitive decline in Alzheimer’s disease. Acta Neuropathol. 2019, 137, 279–296. [Google Scholar] [CrossRef] [Green Version]
  57. Cumba Garcia, L.M.; Peterson, T.E.; Cepeda, M.A.; Johnson, A.J.; Parney, I.F. Isolation and analysis of plasma-derived exosomes in patients with glioma. Front. Oncol. 2019, 9, 651. [Google Scholar] [CrossRef] [Green Version]
  58. Du, Y.; Yu, Y.; Hu, Y.; Li, X.W.; Wei, Z.X.; Pan, R.Y.; Li, X.S.; Zheng, G.E.; Qin, X.Y.; Liu, Q.S.; et al. Genome-wide, integrative analysis implicates exosome-derived microrna dysregulation in schizophrenia. Schizophr. Bull. 2019, 45, 1257–1266. [Google Scholar] [CrossRef] [PubMed]
  59. Gámez-Valero, A.; Campdelacreu, J.; Reñé, R.; Beyer, K.; Borràs, F.E. Comprehensive proteomic profiling of plasma-derived Extracellular Vesicles from dementia with Lewy Bodies patients. Sci. Rep. 2019, 9, 13282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Goetzl, E.J.; Elahi, F.M.; Mustapic, M.; Kapogiannis, D.; Pryhoda, M.; Gilmore, A.; Gorgens, K.A.; Davidson, B.; Granholm, A.C.; Ledreux, A. Altered levels of plasma neuron-derived exosomes and their cargo proteins characterize acute and chronic mild traumatic brain injury. FASEB J. 2019, 33, 5082–5088. [Google Scholar] [CrossRef]
  61. Jain, G.; Stuendl, A.; Rao, P.; Berulava, T.; Pena Centeno, T.; Kaurani, L.; Burkhardt, S.; Delalle, I.; Kornhuber, J.; Hüll, M.; et al. A combined miRNA-piRNA signature to detect Alzheimer’s disease. Transl. Psychiatry 2019, 9, 250. [Google Scholar] [CrossRef] [Green Version]
  62. Jones, P.S.; Yekula, A.; Lansbury, E.; Small, J.L.; Ayinon, C.; Mordecai, S.; Hochberg, F.H.; Tigges, J.; Delcuze, B.; Charest, A.; et al. Characterization of plasma-derived protoporphyrin-IX-positive extracellular vesicles following 5-ALA use in patients with malignant glioma. EBioMedicine 2019, 48, 23–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Ko, J.; Hemphill, M.; Yang, Z.; Beard, K.; Sewell, E.; Shallcross, J.; Schweizer, M.; Sandsmark, D.K.; Diaz-Arrastia, R.; Kim, J.; et al. Multi-dimensional mapping of brain-derived extracellular vesicle MicroRNA biomarker for traumatic brain injury diagnostics. J. Neurotrauma 2019. [Google Scholar] [CrossRef]
  64. Kuharić, J.; Grabušić, K.; Tokmadžić, V.S.; Štifter, S.; Tulić, K.; Shevchuk, O.; Lučin, P.; Šustić, A. Severe traumatic brain injury induces early changes in the physical properties and protein composition of intracranial extracellular vesicles. J. Neurotrauma 2019, 36, 190–200. [Google Scholar] [CrossRef] [PubMed]
  65. Lee, M.Y.; Baxter, D.; Scherler, K.; Kim, T.K.; Wu, X.; Abu-Amara, D.; Flory, J.; Yehuda, R.; Marmar, C.; Jett, M.; et al. Distinct profiles of cell-free MicroRNAs in plasma of veterans with post-traumatic stress disorder. J. Clin. Med. 2019, 8, 963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Lewis, J.; Alattar, A.A.; Akers, J.; Carter, B.S.; Heller, M.; Chen, C.C. A pilot proof-of-principle analysis demonstrating dielectrophoresis (DEP) as a glioblastoma biomarker platform. Sci. Rep. 2019, 9, 10279. [Google Scholar] [CrossRef] [Green Version]
  67. Muraoka, S.; Jedrychowski, M.P.; Tatebe, H.; DeLeo, A.M.; Ikezu, S.; Tokuda, T.; Gygi, S.P.; Stern, R.A.; Ikezu, T. Proteomic profiling of extracellular vesicles isolated from cerebrospinal fluid of former national football league players at risk for chronic traumatic encephalopathy. Front. Neurosci. 2019, 13, 1059. [Google Scholar] [CrossRef] [Green Version]
  68. Otake, K.; Kamiguchi, H.; Hirozane, Y. Identification of biomarkers for amyotrophic lateral sclerosis by comprehensive analysis of exosomal mRNAs in human cerebrospinal fluid. BMC Med. Genomics 2019, 12, 7. [Google Scholar] [CrossRef] [Green Version]
  69. Ricklefs, F.L.; Maire, C.L.; Reimer, R.; Dührsen, L.; Kolbe, K.; Holz, M.; Schneider, E.; Rissiek, A.; Babayan, A.; Hille, C.; et al. Imaging flow cytometry facilitates multiparametric characterization of extracellular vesicles in malignant brain tumours. J. Extracell Vesicles 2019, 8, 1588555. [Google Scholar] [CrossRef] [Green Version]
  70. Saucier, D.; Wajnberg, G.; Roy, J.; Beauregard, A.P.; Chacko, S.; Crapoulet, N.; Fournier, S.; Ghosh, A.; Lewis, S.M.; Marrero, A.; et al. Identification of a circulating miRNA signature in extracellular vesicles collected from amyotrophic lateral sclerosis patients. Brain Res. 2019, 1708, 100–108. [Google Scholar] [CrossRef]
  71. Shao, N.; Xue, L.; Wang, R.; Luo, K.; Zhi, F.; Lan, Q. miR-454-3p is an exosomal biomarker and functions as a tumor suppressor in glioma. Mol. Cancer Ther. 2019, 18, 459–469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Si, X.; Tian, J.; Chen, Y.; Yan, Y.; Pu, J.; Zhang, B. Central nervous system-derived exosomal alpha-synuclein in serum may be a biomarker in Parkinson’s disease. Neuroscience 2019, 413, 308–316. [Google Scholar] [CrossRef]
  73. Wang, H.; Jiang, D.; Li, W.; Xiang, X.; Zhao, J.; Yu, B.; Wang, C.; He, Z.; Zhu, L.; Yang, Y. Evaluation of serum extracellular vesicles as noninvasive diagnostic markers of glioma. Theranostics 2019, 9, 5347–5358. [Google Scholar] [CrossRef] [PubMed]
  74. Winston, C.N.; Romero, H.K.; Ellisman, M.; Nauss, S.; Julovich, D.A.; Conger, T.; Hall, J.R.; Campana, W.; O’Bryant, S.E.; Nievergelt, C.M.; et al. Assessing neuronal and astrocyte derived exosomes from individuals with mild traumatic brain injury for markers of neurodegeneration and cytotoxic activity. Front. Neurosci. 2019, 13, 1005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Banack, S.A.; Dunlop, R.A.; Cox, P.A. An miRNA fingerprint using neural-enriched extracellular vesicles from blood plasma: Towards a biomarker for amyotrophic lateral sclerosis/motor neuron disease. Open Biol. 2020, 10, 200116. [Google Scholar] [CrossRef] [PubMed]
  76. Chen, C.; Wu, Y.; Li, M.; Cui, C.; Zhao, Y.; Sun, X.; Wang, Y.; Liu, C.; Wu, H.; Zhong, X.; et al. Different exosomal microRNA profile in aquaporin-4 antibody positive neuromyelitis optica spectrum disorders. Front. Immunol. 2020, 11, 1064. [Google Scholar] [CrossRef]
  77. Choi, J.; Kim, S.Y.; Kim, H.; Lim, B.C.; Hwang, H.; Chae, J.H.; Kim, K.J.; Oh, S.; Kim, E.Y.; Shin, J.S. Serum α-synuclein and IL-1β are increased and correlated with measures of disease severity in children with epilepsy: Potential prognostic biomarkers? BMC Neurol. 2020, 20, 85. [Google Scholar] [CrossRef]
  78. Devoto, C.; Lai, C.; Qu, B.X.; Guedes, V.A.; Leete, J.; Wilde, E.; Walker, W.C.; Diaz-Arrastia, R.; Kenney, K.; Gill, J. Exosomal MicroRNAs in military personnel with mild traumatic brain injury: Preliminary results from the chronic effects of neurotrauma consortium biomarker discovery project. J. Neurotrauma 2020. [Google Scholar] [CrossRef]
  79. Ebrahimkhani, S.; Beadnall, H.N.; Wang, C.; Suter, C.M.; Barnett, M.H.; Buckland, M.E.; Vafaee, F. Serum exosome MicroRNAs predict multiple sclerosis disease activity after fingolimod treatment. Mol. Neurobiol. 2020, 57, 1245–1258. [Google Scholar] [CrossRef]
  80. Eguchi, A.; Fukuda, S.; Kuratsune, H.; Nojima, J.; Nakatomi, Y.; Watanabe, Y.; Feldstein, A.E. Identification of actin network proteins, talin-1 and filamin-A, in circulating extracellular vesicles as blood biomarkers for human myalgic encephalomyelitis/chronic fatigue syndrome. Brain Behav. Immun. 2020, 84, 106–114. [Google Scholar] [CrossRef]
  81. Goetzl, E.J.; Peltz, C.B.; Mustapic, M.; Kapogiannis, D.; Yaffe, K. Neuron-derived plasma exosome proteins after remote traumatic brain injury. J. Neurotrauma 2020, 37, 382–388. [Google Scholar] [CrossRef]
  82. Goetzl, E.J.; Yaffe, K.; Peltz, C.B.; Ledreux, A.; Gorgens, K.; Davidson, B.; Granholm, A.C.; Mustapic, M.; Kapogiannis, D.; Tweedie, D.; et al. Traumatic brain injury increases plasma astrocyte-derived exosome levels of neurotoxic complement proteins. FASEB J. 2020, 34, 3359–3366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Guedes, V.A.; Kenney, K.; Shahim, P.; Qu, B.X.; Lai, C.; Devoto, C.; Walker, W.C.; Nolen, T.; Diaz-Arrastia, R.; Gill, J.M. Exosomal neurofilament light: A prognostic biomarker for remote symptoms after mild traumatic brain injury? Neurology 2020, 94, e2412–e2423. [Google Scholar] [CrossRef] [PubMed]
  84. Mansur, R.B.; Delgado-Peraza, F.; Subramaniapillai, M.; Lee, Y.; Iacobucci, M.; Rodrigues, N.; Rosenblat, J.D.; Brietzke, E.; Cosgrove, V.E.; Kramer, N.E.; et al. Extracellular vesicle biomarkers reveal inhibition of neuroinflammation by infliximab in association with antidepressant response in adults with bipolar depression. Cells 2020, 9, 865. [Google Scholar] [CrossRef] [Green Version]
  85. Mondello, S.; Guedes, V.A.; Lai, C.; Czeiter, E.; Amrein, K.; Kobeissy, F.; Mechref, Y.; Jeromin, A.; Mithani, S.; Martin, C.; et al. Circulating brain injury exosomal proteins following moderate-to-severe traumatic brain injury: Temporal profile, outcome prediction and therapy implications. Cells 2020, 9, 977. [Google Scholar] [CrossRef] [Green Version]
  86. Nasca, C.; Dobbin, J.; Bigio, B.; Watson, K.; de Angelis, P.; Kautz, M.; Cochran, A.; Mathé, A.A.; Kocsis, J.H.; Lee, F.S.; et al. Insulin receptor substrate in brain-enriched exosomes in subjects with major depression: On the path of creation of biosignatures of central insulin resistance. Mol. Psychiatry 2020. [Google Scholar] [CrossRef] [PubMed]
  87. Nie, C.; Sun, Y.; Zhen, H.; Guo, M.; Ye, J.; Liu, Z.; Yang, Y.; Zhang, X. Differential expression of plasma Exo-miRNA in neurodegenerative diseases by next-generation sequencing. Front. Neurosci. 2020, 14, 438. [Google Scholar] [CrossRef]
  88. Niu, M.; Li, Y.; Li, G.; Zhou, L.; Luo, N.; Yao, M.; Kang, W.; Liu, J. A longitudinal study on α-synuclein in plasma neuronal exosomes as a biomarker for Parkinson’s disease development and progression. Eur. J. Neurol. 2020, 27, 967–974. [Google Scholar] [CrossRef] [PubMed]
  89. Peltz, C.B.; Kenney, K.; Gill, J.; Diaz-Arrastia, R.; Gardner, R.C.; Yaffe, K. Blood biomarkers of traumatic brain injury and cognitive impairment in older veterans. Neurology 2020. [Google Scholar] [CrossRef]
  90. Perrotte, M.; Haddad, M.; Le Page, A.; Frost, E.H.; Fulöp, T.; Ramassamy, C. Profile of pathogenic proteins in total circulating extracellular vesicles in mild cognitive impairment and during the progression of Alzheimer’s disease. Neurobiol. Aging 2020, 86, 102–111. [Google Scholar] [CrossRef]
  91. Puigdelloses, M.; González-Huárriz, M.; García-Moure, M.; Martínez-Vélez, N.; Esparragosa Vázquez, I.; Bruna, J.; Zandio, B.; Agirre, A.; Marigil, M.; Petrirena, G.; et al. RNU6-1 in circulating exosomes differentiates GBM from non-neoplastic brain lesions and PCNSL but not from brain metastases. Neurooncol. Adv. 2020, 2, vdaa010. [Google Scholar] [CrossRef] [Green Version]
  92. Ricklefs, F.L.; Maire, C.L.; Matschke, J.; Dührsen, L.; Sauvigny, T.; Holz, M.; Kolbe, K.; Peine, S.; Herold-Mende, C.; Carter, B.; et al. FASN is a biomarker enriched in malignant glioma-derived extracellular vesicles. Int. J. Mol. Sci. 2020, 21, 1931. [Google Scholar] [CrossRef] [Green Version]
  93. Serpente, M.; Fenoglio, C.; D’Anca, M.; Arcaro, M.; Sorrentino, F.; Visconte, C.; Arighi, A.; Fumagalli, G.G.; Porretti, L.; Cattaneo, A.; et al. MiRNA profiling in plasma neural-derived small extracellular vesicles from patients with Alzheimer’s disease. Cells 2020, 9, 1443. [Google Scholar] [CrossRef]
  94. Tabibkhooei, A.; Izadpanahi, M.; Arab, A.; Zare-Mirzaei, A.; Minaeian, S.; Rostami, A.; Mohsenian, A. Profiling of novel circulating microRNAs as a non-invasive biomarker in diagnosis and follow-up of high and low-grade gliomas. Clin. Neurol. Neurosurg. 2020, 190, 105652. [Google Scholar] [CrossRef]
  95. Tan, N.; Hu, S.; Hu, Z.; Wu, Z.; Wang, B. Quantitative proteomic characterization of microvesicles/exosomes from the cerebrospinal fluid of patients with acute bilirubin encephalopathy. Mol. Med. Rep. 2020, 22, 1257–1268. [Google Scholar] [CrossRef] [PubMed]
  96. Wang, D.; Wang, P.; Bian, X.; Xu, S.; Zhou, Q.; Zhang, Y.; Ding, M.; Han, M.; Huang, L.; Bi, J.; et al. Elevated plasma levels of exosomal BACE1-AS combined with the volume and thickness of the right entorhinal cortex may serve as a biomarker for the detection of Alzheimer’s disease. Mol. Med. Rep. 2020, 22, 227–238. [Google Scholar] [CrossRef] [PubMed]
  97. Galbo, P.M., Jr.; Ciesielski, M.J.; Figel, S.; Maguire, O.; Qiu, J.; Wiltsie, L.; Minderman, H.; Fenstermaker, R.A. Circulating CD9+/GFAP+/survivin+ exosomes in malignant glioma patients following survivin vaccination. Oncotarget 2017, 8, 114722–114735. [Google Scholar] [CrossRef] [PubMed]
  98. Masters, C.L.; Bateman, R.; Blennow, K.; Rowe, C.C.; Sperling, R.A.; Cummings, J.L. Alzheimer’s disease. Nat. Rev. Dis. Primers 2015, 1, 15056. [Google Scholar] [CrossRef] [PubMed]
  99. Petersen, R.C. Clinical practice. Mild cognitive impairment. N. Engl. J. Med. 2011, 364, 2227–2234. [Google Scholar] [CrossRef] [Green Version]
  100. Poewe, W.; Seppi, K.; Tanner, C.M.; Halliday, G.M.; Brundin, P.; Volkmann, J.; Schrag, A.E.; Lang, A.E. Parkinson disease. Nat. Rev. Dis. Primers 2017, 3, 17013. [Google Scholar] [CrossRef]
  101. Filippi, M.; Bar-Or, A.; Piehl, F.; Preziosa, P.; Solari, A.; Vukusic, S.; Rocca, M.A. Multiple sclerosis. Nat. Rev. Dis. Primers 2018, 4, 43. [Google Scholar] [CrossRef] [PubMed]
  102. Hardiman, O.; Al-Chalabi, A.; Chio, A.; Corr, E.M.; Logroscino, G.; Robberecht, W.; Shaw, P.J.; Simmons, Z.; van den Berg, L.H. Amyotrophic lateral sclerosis. Nat. Rev. Dis. Primers 2017, 3, 17071. [Google Scholar] [CrossRef] [PubMed]
  103. Mattick, J.S.; Rinn, J.L. Discovery and annotation of long noncoding RNAs. Nat. Struct. Mol. Biol. 2015, 22, 5–7. [Google Scholar] [CrossRef] [PubMed]
  104. Ramalho-Carvalho, J.; Fromm, B.; Henrique, R.; Jeronimo, C. Deciphering the function of non-coding RNAs in prostate cancer. Cancer Metastasis Rev. 2016, 35, 235–262. [Google Scholar] [CrossRef] [PubMed]
  105. Louis, D.N.; Perry, A.; Reifenberger, G.; von Deimling, A.; Figarella-Branger, D.; Cavenee, W.K.; Ohgaki, H.; Wiestler, O.D.; Kleihues, P.; Ellison, D.W. The 2016 World Health Organization classification of tumors of the central nervous system: A summary. Acta Neuropathol. 2016, 131, 803–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Blennow, K.; Brody, D.L.; Kochanek, P.M.; Levin, H.; McKee, A.; Ribbers, G.M.; Yaffe, K.; Zetterberg, H. Traumatic brain injuries. Nat. Rev. Dis. Primers 2016, 2, 16084. [Google Scholar] [CrossRef] [PubMed]
  107. Levin, H.S.; Diaz-Arrastia, R.R. Diagnosis, prognosis, and clinical management of mild traumatic brain injury. Lancet Neurol. 2015, 14, 506–517. [Google Scholar] [CrossRef]
  108. Marder, S.R.; Cannon, T.D. Schizophrenia. N. Engl. J. Med. 2019, 381, 1753–1761. [Google Scholar] [CrossRef]
  109. Collaborators, G.B.D.S. Global, regional, and national burden of stroke, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 439–458. [Google Scholar] [CrossRef] [Green Version]
  110. Campbell, B.C.V.; De Silva, D.A.; Macleod, M.R.; Coutts, S.B.; Schwamm, L.H.; Davis, S.M.; Donnan, G.A. Ischaemic stroke. Nat. Rev. Dis. Primers 2019, 5, 70. [Google Scholar] [CrossRef]
  111. Xu, X.; Jiang, Y. The Yin and Yang of innate immunity in stroke. Biomed. Res. Int. 2014, 2014, 807978. [Google Scholar] [CrossRef] [Green Version]
  112. Brand, F.J., 3rd; de Rivero Vaccari, J.C.; Mejias, N.H.; Alonso, O.F.; de Rivero Vaccari, J.P. RIG-I contributes to the innate immune response after cerebral ischemia. J. Inflamm. 2015, 12, 52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Neumann, S.; Shields, N.J.; Balle, T.; Chebib, M.; Clarkson, A.N. Innate immunity and inflammation post-stroke: An alpha7-nicotinic agonist perspective. Int. J. Mol. Sci. 2015, 16, 29029–29046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Otte, C.; Gold, S.M.; Penninx, B.W.; Pariante, C.M.; Etkin, A.; Fava, M.; Mohr, D.C.; Schatzberg, A.F. Major depressive disorder. Nat. Rev. Dis. Primers 2016, 2, 16065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Kupfer, D.J.; Frank, E.; Phillips, M.L. Major depressive disorder: New clinical, neurobiological, and treatment perspectives. Lancet 2012, 379, 1045–1055. [Google Scholar] [CrossRef] [Green Version]
  116. Etkin, A.; Buchel, C.; Gross, J.J. The neural bases of emotion regulation. Nat. Rev. Neurosci. 2015, 16, 693–700. [Google Scholar] [CrossRef] [PubMed]
  117. Watson, K.; Nasca, C.; Aasly, L.; McEwen, B.; Rasgon, N. Insulin resistance, an unmasked culprit in depressive disorders: Promises for interventions. Neuropharmacology 2018, 136, 327–334. [Google Scholar] [CrossRef]
  118. Nasca, C.; Rasgon, N.; McEwen, B. An emerging epigenetic framework of systemic and central mechanisms underlying stress-related disorders. Neuropsychopharmacology 2019, 44, 235–236. [Google Scholar] [CrossRef] [Green Version]
  119. Duarte, A.I.; Moreira, P.I.; Oliveira, C.R. Insulin in central nervous system: More than just a peripheral hormone. J. Aging Res. 2012, 2012, 384017. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Flow diagram representing a summary of the conducted methodology for this review.
Figure 1. Flow diagram representing a summary of the conducted methodology for this review.
Ijms 22 03267 g001
Figure 2. Schematic representation of the brain microenvironment, its main cellular components and how the brain–blood-barrier permeability allows for extracellular vesicles to reach circulation. Created with BioRender.com.
Figure 2. Schematic representation of the brain microenvironment, its main cellular components and how the brain–blood-barrier permeability allows for extracellular vesicles to reach circulation. Created with BioRender.com.
Ijms 22 03267 g002
Figure 3. Schematic representation of the use of extracellular vesicles in liquid biopsies for identification of specific central nervous system (CNS)-related pathological conditions. Created with BioRender.com (accessed on 20 March 2021).
Figure 3. Schematic representation of the use of extracellular vesicles in liquid biopsies for identification of specific central nervous system (CNS)-related pathological conditions. Created with BioRender.com (accessed on 20 March 2021).
Ijms 22 03267 g003
Table 1. Extracellular vesicles protein and protein-coding genes transcripts as biomarkers associated with central nervous system pathological conditions.
Table 1. Extracellular vesicles protein and protein-coding genes transcripts as biomarkers associated with central nervous system pathological conditions.
MoleculeDiseaseSample TypeSample Grouping and SizePatients Gender and Mean Age EVs Isolation MethodKey FindingsRef.
pSer312,
p-panTyr-IRS-1
ADPlasmaPatients
(N = 24)
14♀ + 10♂ (73 yrs)Thromboplastin-D + ExoQuick® (System Biosciences)
+ L1CAM IP
Markers of brain insulin resistance in NDEVs associate with atrophy in AD.[29]
NPTX2,
NRXN2a,
AMPA4,
NLGN1
ADPlasmaC.S.S.:
Patients (N = 28),
HC (N = 28);
L.S.:
Patients (N = 18 + 18), HC (N = 18)
C.S.S.: Patients and HC—16♀ + 12♂ (73 yrs);
L.S.:
Patients and HC—8♀ + 10♂ (69 & 78 yrs)
Thromboplastin-D + 3000× g (30′ at 4 °C) + ExoQuick®
+ L1CAM IP
Reduction of the marker levels in NDEVs may be indicative of the extent of cognitive loss and reflect progression of the severity of AD.[35]
TauADPlasmaAD patients (N = 20),
MCI patients (N = 10),
HC (N = 10)
AD—11♀ + 9♂ (75 yrs)
MCI—5♀ + 5♂ (76 yrs)
HC—7♀ + 3♂ (76 yrs)
Thrombin + 6000× g (20′ at 4 °C) + ExoQuick®
+ L1CAM IP
Tau was found free-floating with a small component inside EVs; full-length Tau was higher inside EVs than in free solution. [36]
SNAP-25ADSerumAD patients (N = 24),
HC (N = 17)
AD—16♀ + 8♂ (78 yrs)
HC—13♀ + 4♂ (77 yrs)
10,000× g (10′ at RT) + ExoQuick®
+ L1CAM IP
The levels of SNAP-25 carried by NDEVs were reduced in AD patients (sensitivity 87.5%, specificity 70.6%).[53]
N-123 tau,
N-224 tau
ADSerumPatients w/CSF+ biomarkers (N = 4), patients w/CSF- biomarkers (N = 4) n.m.4000× g (20′ at 4 °C) + ExoQuick®
+ L1CAM IP
N-224 tau was present in NDEVs, while N-123 tau showed comparable concentrations in both NDEVs and peripherally derived EVs. [56]
BACE1-ASADPlasmaAD patients (N = 72),
HC (N = 62)
AD—38♀ + 34♂
(74 yrs)
HC—39♀ + 23♂
(72 yrs)
Thrombin + 14,000× g (5′ at 4 °C) + 3000× g (15′ at 4 °C) + ExoQuick®EVs BACE1-AS transcript levels in AD patients were significantly higher compared with the HC (sensitivity 87.5%, specificity 61.3%) [96]
SYP,
SYNPO,
SYT2,
NRGN,
GAP43,
SYN1
AD and FTDPlasmaC.S.S.:
AD patients (N = 12),
FTD patients (N = 16), HC (N = 28);
L.S.:
AD patients (N = 9), FTD patients (N = 10),
HC (N = 19)
C.S.S.:
AD—6♀ + 6♂ (74 yrs)
FTD—4♀ + 12♂ (64 yrs)
L.S.:
AD—7♀ + 2♂ (82 yrs)
FTD—5♀ + 5♂ (63 yrs)
Thromboplastin-D + 3000× g (30′ at 4 °C) + ExoQuick®
+ L1CAM IP
SYP, SYNPO, SYT2, and NRGN levels were significantly lower in patients with FTD and AD than in HC. Some markers were decreased years before dementia in FTD and AD patients. [22]
TauAD and PDPlasmaAD patients (N = 106),
PD patients (N = 91),
HC (N = 106)
AD—49♀ + 57♂ (70 yrs)
PD—26♀ + 65♂ (65 yrs)
HC—48♀ + 58♂ (67 yrs)
2000× g (15′) + ultracentrifugation + L1CAM IPTau was significantly higher in PD patients than HCs, but not in AD patients, and correlated
with CSF tau.
[25]
pS1292-LRRK2PDCSFPD patients:
LRRK2+ mutation (N = 19),
LRRK2− (N = 19);
HC:
LRRK2+ (N = 39),
LRRK2- (N = 5)
PD patients:
LRRK2+
16♀ + 3♂ (57 yrs)
LRRK2−
8♀ + 11♂ (60 yrs)
HC: LRRK2+
26♀ + 13♂ (63 yrs)
LRRK2- 5♀ (60 yrs)
10,000× g (30′ at 4 °C) +
ultracentrifugation
pS1292-LRRK2 levels in CSF EVs were near saturated in most subjects, 10-fold higher than in urinary EVs, irrespective of LRRK2 mutation status or PD diagnosis.[30]
Akt,
p-mTOR,
p-Tyr-IRS-1
PDSerumPD patients (N = 60)17♀+ 43♂ (60 yrs)4500× g (20′ at 4 °C) + ExoQuick®
+ L1CAM IP
Exenatide-treated patients had elevated expression of tyrosine phosphorylation of IRS-1 and of downstream targets, total Akt and p-mTOR.[54]
α-synucleinPDPlasmaPD patients (N = 267), HC (N = 215)PD—119♀ + 145♂ (66 yrs)
HC—99♀ + 116♂ (66 yrs)
2000× g (15′) + ultracentrifugation + L1CAM IPLevels of α-synuclein in EVs were substantially higher in PD patients than in HC. A significant correlation between α-synuclein found on EVs and disease severity was observed.[18]
α-synucleinPDSerumPD patients:
Tremor type (N = 22),
Non-tremor (N = 16);
essential tremor (ET) patients (N = 21);
HC (N = 18)
Tremor type—10♀ + 12♂ (63 yrs)
Non-tremor type—9♀ + 9♂ (62 yrs)
ET—10♀ + 11♂ (62 yrs)
HC—10♀ + 8♂ (63 yrs)
3000× g for (15′ at 4 °C) + ExoQuick®
+ L1CAM IP
α-synuclein levels were lower in the PD group than in the ET and HC. Levels were lower in the NTD group than in the TD group. α-synuclein was found to moderately aid in PD diagnosis (AUC = 0.675) and had a potential to diagnose NTD (AUC = 0.761).[72]
α-synucleinPDPlasmaEarly-stage PD patients (N = 36),
Advanced PD patients (N = 17),
iRBD patients (N = 20), HC (N = 21)
Early-stage PD—18♀ + 18♂ (64 yrs)
Advanced PD—10♀ + 7♂ (67 yrs)
iRBD—8♀ + 12♂ (63 yrs)
HC—10♀ + 11♂ (64 yrs)
2000× g (15′) + ultracentrifugation + L1CAM IPα-synuclein levels in NDEVs were significantly higher in patients with early-stage PD compared with HCs. Longitudinally increased α-synuclein were associated with higher risk for motor symptom progression in PD.[88]
Aß1-42,
p-Tau-S396,
NRGN,
SYP,
SYT1,
SYNPO
MCIPlasmaMCI patients (N = 61),
HC (N = 76)
MCI—39♀ + 22♂ (70 yrs)
HC—47♀ + 29♂ (68 yrs)
Thrombin + 10,000 rpm (5′) +
ExoQuick®
+ L1CAM IP
NDEVs concentrations of Aβ1-42 were significantly increased while NRGN, synaptophysin, synaptotagmin, and synaptopodin levels were significantly decreased in patients with MCI.[52]
apoA1,
apoE,
apoJ,
AnnexinV,
Aß-42
MCI, AD and
PD
CSFMCI patients (N = 21),
AD patients (N = 27),
PD patients (N = 28),
young HC (N = 15),
middle-aged HC (N = 21),
older HC (N = 23)
MCI—7♀ + 14♂ (75 yrs)
AD—11♀ + 16♂ (69 yrs)
PD—12♀ + 12♂ (64 yrs)
Young HC—11♀ + 4♂ (28yrs)
Middle-aged HC—10♀ + 11♂ (55 yrs)
Older HC—12♀ + 11♂ (73yrs)
Flow cytometric assayApoE and Aß-42-positive particle concentrations were reduced in middle and older age subjects, whereas apoAI increased with age. ApoAI and annexin V levels were reduced in MCI and/or AD patients vs. HCs.[21]
GSN,
BCHE
DLBPlasmaDLB patients (N = 19),
AD patients (N = 10),
HC (N = 20)
DLB—ratio ♀/♂ 2:3 (72 yrs)
AD—ratio ♀/♂ 2:3 (74 yrs)
HC—ratio ♀/♂ 2:1 (69 yrs)
2500× g (15′) + 16,000× g (10′) + SECGelsolin decreased levels were found on EVs from DLB patients, compared to HCs and to AD patients.[59]
P-tau,
Aß1-42,
NRGN,
REST
MCI and ADPlasmaMCI patients (N = 20),
AD patients (N = 10),
MCI-to-AD (ADC) patients (N = 20)
HC (N = 10)
MCI—7♀ + 13♂ (69 yrs)
ADC—9♀ + 11♂ (75 yrs)
AD and HC—n.m.
Thromboplastin-D + ExoQuick®
+ L1CAM IP
Abnormal NDEVs levels of P-tau, Aß1-42, NRGN and REST accurately predicted conversion of MCI to AD dementia. [26]
Tau,
APP,
pTau-T181,
Aβ42
MCI and ADPlasmaMCI patients (N = 12),
mild AD patients (N = 12),
moderate AD patients (N = 12),
severe AD patients (N = 20),
HC (N = 12)
MCI—11♀ + 1♂ (75 yrs)
Mild AD—11♀ + 1♂ (76yrs)
Moderate AD patients
8♀ + 4♂ (79 yrs)
Severe AD patients
10♀ + 2♂ (83 yrs)
HC—9♀ + 3♂ (69 yrs)
2000× g (20′) + 10,000× g (20′) + Total Exosome Isolation reagent
(InvitrogenTM)
Abnormal APP levels and pTau-T181/tTau ratio in EVs demonstrated a high accuracy to define MCI and AD staging.[90]
FN1,
GFAP
NMOSDCSFMS patients (N = 10),
NMOSD patients (N = 10),
idiopathic longitudinally extensive transverse myelitis patients (N = 12)
MS—7♀ + 3♂ (n.m.)
NMO—9♀ + 1♂ (n.m.)
I-LETM—1♀ + 11♂ (n.m.)
18,000× g (30′) +
ultracentrifugation
442 significant proteins generated a list of signature molecules of diseases validated primarily by the identification of known markers such as GFAP and FN1, specific to NMO and MS.[24]
KLKB1,
APOE
MSCSFRRMS patients (N = 4),
non-demyelinating controls (N = 3)
n.m.Exo-SpinTM (Cell Guidance Systems)Plasma kallikrein and Apolipoprotein-E4 were increased in CSF-EVs compared to CSF.[31]
MOGMSSerum and CSFRRMS patients (N = 45),
secondary progressive MS (SPMS) patients (N = 30),
HC (N = 45)
n.m.ExoQuick®Exosomal content of MOG strongly correlated with disease activity and was highest in RRMS patients in relapse and in SPMS patients.[34]
ASMaseMSCSFMS patients (N = 95),
other central neurological disease (C_OND) patients (N = 45),
other peripheral neurological disease (P_OND) patients (N = 31)
MCI—55♀ + 40♂ (37yrs)
C_OND—29♀ + 16♂ (43yrs)
P_OND—13♀ + 18♂ (58yrs)
Flow cytometric assayA high number of acid sphingomyelinase-enriched EVs correlated to enzymatic activity and to disease severity.[43]
SOD1,
TDP-43,
p-TDP-43,
FUS
ALSPlasmaALS patients (N = 30)
HC (N = 30)
ALS—15♀ + 15♂ (71 yrs)
HC – n.m.
1600× g (20′) +
ultracentrifugation
Microvesicles (MVs) and Exosomes (EXOs) size were increased in ALS patients compared to HCs; MVs of ALS patients were enriched with toxic proteins compared to HCs.[48]
CUEDC2ALSCSFALS patients (N = 4),
HC (N = 4)
ALS—4♂ (58 yrs)
HC—4♂ (59 yrs)
2000× g (5′ at 4 °C) + 10,000× g (20′ at 4 °C) + ExoRNeasy Serum/Plasma Midi Kit (QIAGEN)By RNA sequencing, several genes, such as CUEDC2, in CSF EVs were suggested to be candidate disease biomarkers for ALS.[68]
CD14,
Cystatin C
CWML/Brain atrophyPlasmaManifest vascular disease patients (N = 994) 210♀ + 784♂ (59 yrs)3000× g (15′) + ExoQuick®EV proteins cystatin C and CD14 were related to CWMLs and the progression of brain atrophy in patients with manifest vascular disease.[16]
Ras-related small GTPase 10,
Annexin VII,
UCHL1,
Claudin-5,
NKCC1,
AQP4,
SYNGR3,
Aβ42,
P-T181-tau,
P-S396-tau,
IL-6,
PRPc
TBIPlasmaAcute mild TBI (N = 18),
chronic mild TBI (N = 14),
HC (N = 21)
Acute mild TBI—12♀ + 6♂ (21 yrs)
Chronic mild TBI—3♀ + 11♂ (20 yrs)
HC—14♀ + 7♂ (21 yrs)
Thromboplastin-D + 3000× g (30′ at 4 °C) + ExoQuick®
+ L1CAM IP
Increases in NDEV levels of most neurofunctional proteins in acute mild TBI, and elevations of most NDEV neuropathological proteins in chronic and acute mild TBI delineated phase-specificity.[60]
FLOT1,
Arf6,
Rab7a
TBICSFSevere TBI patients (N = 17),
HC (N = 18)
Severe TBI—2♀ + 15♂ (40 yrs)
HC—n.m.
500× g (10′ at 4 °C) + 2000× g (30′ at 4 °C) + ultracentrifugationCSF after severe TBI contains Flotillin+ EVs. Unfavorable outcomes included decreasing Arf6 concentrations and a delayed Rab7a concentration increase.[64]
Aβ42,
NRGN
TBIPlasmaMild TBI patients (N = 19),
HC (N = 20)
Mild TBI—19♂ (22 yrs)
HC—20♂ (22 yrs)
Thrombin + 10,000 rpm (5′) +
ExoQuick® + L1CAM or
GLAST IP
NDEV and ADEV levels of Aβ42 were significantly higher while NDEV and ADEV levels of neurogranin were significantly lower in mild TBI patients compared to HCs.[74]
Aβ42,
P-tau,
PRPc,
SYNGR3
TBIPlasmaTBI patients:
W/CI (N = 26),
W/o CI (N = 21);
Controls:
W/CI (N = 19),
W/o CI (N = 42)
TBI w/CI—26♂ (75 yrs)
TBI w/o CI—3♀ + 18♂ (79 yrs)
Controls w/CI—1♀ + 18♂ (80 yrs)
Controls w/o CI—7♀ + 35♂ (79 yrs)
Thromboplastin-D + 3000× g (30′ at 4 °C) + ExoQuick®
+ L1CAM IP
Aβ42 and P-tau species, and their respective putative receptors, PrPc and synaptogyrin-3, remain elevated for decades after TBI, and may mediate TBI-associated CI.[81]
Complement effector/regulatory proteinsTBIPlasmasTBI patients (N = 24);
mtTBI patients:
Early (N = 10) and
late (N = 15);
sTBI controls (N = 12);
mtTBI controls:
Early (N = 5) and
late (N = 5)
sTBI—12♀ + 12♂ (21 yrs)
Early mtTBI—3♀ + 7♂ (38 yrs)
Late mtTBI—2♀ + 13♂ (77 yrs)
sTBI controls—6♀ + 6♂ (22 yrs)
Early mtTBI controls—1♀ + 4♂ (38 yrs)
Late mtTBI controls—2♀ + 3♂ (75 yrs)
Thromboplastin-D + 3000× g (30′ at 4 °C) + ExoQuick®
+ GLAST IP
TBI increased plasma ADEVs levels of neurotoxic complement proteins.[82]
NfLTBIPlasma1–2 TBIs patients (N = 94),
≥3 TBIs patients (N = 56),
HC (N = 45)
1–2 TBIs—12♀ + 82♂ (38 yrs)
≥3 TBIs—9♀ + 47♂ (37 yrs)
HC—7♀ + 38♂ (38 yrs)
Thrombin + 10,000 rpm (5–10′) + ExoQuick® Repetitive mild TBIs were associated with elevated EV levels of NfL, even years following these injuries.[83]
UCH-L1,
GFAP,
NfL,
Tau
TBISerumTBI patients (N = 21)3♀ + 18♂ (52 yrs)3000× g (15′ at 4 °C) + ExoQuick®Patients with diffuse injury displayed higher acute EVs NFL and GFAP concentrations than those with focal lesions. EVs UCH-L1 specific profile was associated with early mortality.[85]
NfL,
GFAP,
p-Tau,
TNFa,
IL-6
TBIPlasmaTBI patients:
W/CI (N = 35),
W/o CI (N = 30);
Controls:
W/CI (N = 30),
W/o CI (N = 60)
TBI w/CI—35♂ (77 yrs)
TBI w/o CI—4♀ + 26♂ (80 yrs)
Controls w/CI—3♀ + 27♂ (82 yrs)
Controls w/o CI—9♀ + 51♂ (79 yrs)
Thrombin + ExoQuick®
+ L1CAM IP
All significantly associated biomarkers combined separated TBI w/vs. w/o CI (AUC = 0.85) and CI w/vs. w/o TBI (AUC = 0.88).[89]
ASC,
caspase-1,
IL-1β,
IL-18
StrokeSerumPatients (N = 16),
HC (N = 80)
Stroke—n.m.
HC—40♀ + 40♂ (n.m.)
2000× g (30′) +
Total Exosome Isolation reagent/
3000× g (15′) + ExoQuick®
The AUC for ASC was 0.99, whereas the AUC for caspase-1, IL-1β, and IL-18 were 0.75, 0.61, and 0.67, respectively, and can act as biomarkers for stroke.[38]
Tau,
p-tau181
Chronic traumatic encephalopathyCSFPatients (N = 15),
HC (N = 16)
Patients—15♂ (n.m.)
HC—16♂ (n.m.)
1200× g (20′ at 4 °C) + 10,000× g (30′ at 4 °C) + MagCaptureTM Exosome Isolation Kit PS (FUJIFILM Wako Pure Chemical Corporation)T-tau and p-tau181 levels of CSF-derived EV were positively correlated with the t-tau and p-tau181 levels of total CSF in patients, respectively, but not in the HCs. [67]
TLN1,
FLNA,
14-3-3 proteins
ME/CFSPlasmaME/CFS patients (N = 99),
ICF patients (N = 6),
depression patients (N = 8),
HC (N = 56)
n.m.SEC (qEV iZON Science)Talin-1, filamin-A, and 14-3-3 family proteins were the most abundant proteins in EVs from ME/CFS patients.[80]
S100a9,
S100a7,
lTF,
DEFA1
ABECSFModerate ABE patients (N = 10),
severe ABE patients (N = 10),
HC (N = 10)
Moderate ABE—4♀ + 6♂ (5.7 days)
Severe ABE—3♀ + 7♂ (5.5 days)
HC—5♀ + 5♂ (6.4 days)
2000× g (20′ at 4 °C) + Ribo™ exosome isolation reagent A total of 291 dysregulated proteins were identified by comparing ABE patients with HCs, by mass spectrometry. S100a9, S100a7, lTF and DEFA1 were further validated.[95]
α-synuclein,
IL-1β
Epilepsy and ADDSerumEpilepsy patients (N = 115),
ADD patients (N = 10),
HC (N = 146)
Epilepsy—47♀ + 68♂ (9 yrs)
ADD—7♀ + 3♂ (8 yrs)
HC—68♀ + 76♂ (9 yrs)
3000× g (15′ at 4 °C) + ExoQuick®α-synuclein levels were significantly increased in children with epilepsy and with ADD of the CNS and correlated with measures of disease severity. IL-1β levels showed significant correlation only with drug resistance in children with epilepsy.[77]
PhosphatidylserineSCZCSFSCZ patients (N = 2),
HC (N = 14)
SCZ—2♀ (56 yrs)
HC—n.m.
2000× g (20′ at RT) + 13,000× g (2′ at RT) + flow cytometric assaySCZ patients displayed more phosphatidylserine+ EVs in CSF compared with HCs.[15]
IL-34,
SYP,
TNFR1
MDDPlasmaMDD patients (N = 34),
HC (N = 34)
MDD—14♀ + 20♂ (31 yrs)
HC—14♀ + 20♂ (n.m.)
“Sandwich” ELISA w/CD81 IL-34/CD81 levels were significantly higher in MDD group compared to HC group. Synaptophysin (SYP), SYP/CD81, and TNFR1/CD81 were positively correlated with severities of depression and/or various subsymptoms.[39]
IRS-1MDDPlasmaMDD patients (N = 64),
HC (N = 29)
MDD—40♀ + 24♂ (43 yrs)
HC—11♀ + 18♂
(38 yrs)
Thrombin + 4500× g (20′ at 4 °C) + ExoQuick® + L1CAM IPAn increased concentration of IRS-1 in EVs of MDD patients was found, as compared with HC. Gender differences were observed for serine-312 phosphorylation of IRS-1 in MDD patients EVs.[86]
TNFR1,
NF-κB
Bipolar disorderPlasmaPatients:
Infliximab-treated (N = 27),
placebo (N = 28)
Infliximab—20♀ + 7♂ (44 yrs)
Placebo—24♀ + 4♂ (46 yrs)
Thrombin + 4500× g (20′ at 4 °C) + ExoQuick®
+ L1CAM IP
Higher levels of physical abuse were associated with larger biomarker decreases over time. The antidepressant response to infliximab was moderated by TNFR1. In infliximab-treated participants, reductions in TNFR1 levels were associated with improvement in depressive symptoms.[84]
MGMT,
APNG
GMSerumGM patients (N = 17),
HC (N = 15)
n.m. 1100× g (10′) + immunomagnetic exosomal RNA (iMER) platformEVs mRNA levels of MGMT and APNG correlate well with levels found in parental cells and change considerably during treatment of seven GM patients.[19]
EGFRvIII mutationGMCSFGM patients (N = 71)20♀ + 51♂ (61 yrs)1500× g (10′) +
ultracentrifugation
EGFRvIII was detected in CSF-derived EVs for 14/23 EGFRvIII tissue+ GM patients. Only one of the 48 EGFRvIII tissue- patients had the EGFRvIII mutation detected in EVs. Sensitivity and specificity of EVs to detect an EGFRvIII-positive GBM was 61% and 98%, respectively. [28]
EGFRvIII mutationGMSerum/plasmaGM patients (N = 13),
HC (N = 6)
GM—4♀ + 8♂ (63 yrs)
HC—n.m.
Microfluidic isolationThe EVHB-Chip achieved 94% tumor-EV specificity. EVs from serum and plasma samples from GM patients had mutant EGFRvIII mRNA.[44]
PD-L1GMSerum/plasmaGM patients (N = 21),
HC (N = 5)
n.m.15,000× g (10′) +
ultracentrifugation
PD-L1 DNA was present in circulating EVs from GM patients where it correlated with tumor volumes of up to 60cm3.[45]
IFN-γ,
IL-10,
IL-3,
B7-1,
B7-2,
ICOSL
GMPlasmaGM patients (N = 19),
HC (N = 19)
GM—6♀ + 13♂ (n.m.)
HC—n.m.
3000 rpm (15′) +
OptiPrepTM solution (Sigma-Aldrich) + ultracentrifugation
Cytokines and costimulatory molecules were readily detected but appeared globally reduced in GM patients’ EVs.[57]
GFAP,
Tau
GMPlasmaGM patients (N = 15),
HC (N = 8)
n.m.Dielectrophoretic (DEP) micro-chip deviceFor GM diagnosis, EV-GFAP reached 93% sensitivity, 38% specificity, and AUC of 0.65; for EV-Tau, 67% sensitivity, 75% specificity and AUC of 0.71 was disclosed.[66]
PTRFGliomaSerumGlioma patients (N = 36)n.m.10,000× g (30′) + ultracentrifugationA positive correlation between tumor grade and PTRF expression was found in both tumor tissues and blood EVs from GM patients. PTRF expression in exosomes isolated from the sera of GM patients was decreased after surgery.[37]
EGFRvIII mutationGliomaSerumGrade III glioma patients (N = 23),
Grade IV glioma patients (N = 73)
Other neurological diseases patients (N = 15),
HC (N = 50)
Grade III glioma—4♀ + 19♂ (44 yrs)
Grade IV glioma—25♀ + 48♂ (53 yrs)
Controls—n.m.
600× g (10′) + 2000× g (20′) + 10,000× g (20′) +
Total Exosome Isolation Kit
Sensitivity and specificity of EVs EGFRvIII detection assay in serum were 81.58% and 79.31%, respectively. EGFRvIII expression either in EVs or tissue correlated with poor survival.[41]
EGFR,
NLGN3,
PTTG1
GliomaSerumGlioma patients (N = 23),
HC (N = 12)
Glioma—9♀ + 14♂ (52 yrs)
HC—3♀ + 9♂ (59 yrs)
2000× g (15′) + ultracentrifugationProtein expression of EGFR in EVs can accurately differentiate high-grade and low-grade glioma patients, and positively correlates with ki-67 labeling index in tumor tissue. NLGN3 and PTTG1 mRNA in EVs were also validated for detecting glioma patients.[73]
FASNGliomaPlasmaGlioma patients (N = 8 + 9),
HC (N = 8 + 3)
n.m.1000× g (7′) + 10,000× g (30′) +
ultracentrifugation
FASN was elevated in CD63+ and CD81+ EVs in glioma patient samples.[92]
GFAP,
Survivin
GliomaSerumGlioma patients (N = 8),
HC (N = 3)
Glioma—3♀ + 5♂ (52 yrs)
HC—n.m.
10,000× g (80′at 4 °C) +
ultracentrifugation
Patients with longer time to tumor progression exhibited a decrease in CD9+/SVN+ and CD9+/GFAP+/SVN+ EVs immediately following survivin vaccination; whereas, those with early tumor progression had an increase in the same markers, despite anti-survivin immunotherapy.[97]
PpIXGliomaPlasmaGlioma patients (N = 6)2♀ + 4♂ (59 yrs)exoEasy Maxi Kit (Qiagen)Plasma of patients with avidly fluorescent tumors undergoing FGS contain circulating PpIX+ EVs at levels significantly higher than their predosing background, which correlates with enhancing tumor volumes.[62]
CD63,
CD81
Brain tumors
(mixed)
PlasmaGM patients (n.m.),
anaplastic astrocytoma patients (n.m.),
brain metastases patients (n.m.),
meningioma patients (n.m),
Pituitary adenoma patients (n.m.),
epilepsy controls (n.m.),
HC (n.m.)
n.m.15,000× g (15′) +
ultracentrifugation
EVs with double positive CD63+/CD81+ expression are enriched in cancer cell lines and patient plasma samples. [69]
Abbreviations: ABE—acute bilirubin encephalopathy; AD—Alzheimer’s disease; ADD—acquired demyelinating disorders; ADEV—astrocyte-derived extracellular vesicles; ALS—amyotrophic lateral sclerosis; CI—cognitive Impairment; CSF—cerebrospinal fluid; CWML—cerebral white matter lesion; DLB—dementia with Lewy bodies; EV—extracellular vesicles; FTD—frontotemporal dementia; GM—glioblastoma multiforme; HC—healthy controls; ICF—idiopathic chronic fatigue; MCI—mild cognitive impairment; MDD—major depressive disorder; ME/CFS—myalgic encephalomyelitis/chronic fatigue syndrome; miR—microRNA; MS—multiple sclerosis; mtTBI—military veterans traumatic brain injury; NDEV—neuron-derived extracellular vesicles; n.m.—not mentioned; NMOSD—neuromyelitis optica spectrum disorders; PD—Parkinson’s disease; RRMS—relapsing-remitting multiple sclerosis; SCZ—schizophrenia; SEC—size exclusion chromatography; sTBI—sports-related traumatic brain injury; TBI—traumatic brain injury; yrs—years.
Table 2. Extracellular vesicles non-coding RNAs as biomarkers associated with central nervous system pathological conditions.
Table 2. Extracellular vesicles non-coding RNAs as biomarkers associated with central nervous system pathological conditions.
MoleculeDiseaseSample TypeSample Grouping and SizePatients Gender and Mean Age EVs Isolation MethodKey FindingsRef.
miR-16-5p,
miR-125b-5p,
miR-451a,
miR-605-5p
ADCSFYoung-onset AD (YOAD) Patients (N = 17),
Late-onset AD (LOAD) Patients (N = 13)
HC (N = 12)
YOAD—10♀ + 7♂ (61 yrs)
LOAD—5♀ + 8♂ (76 yrs)
HC—7♀ + 5♂ (67 yrs)
3000× g (5′) +
miRCURYTM Exosome Isolation Kit (Exiqon)
MiR-16-5p, miR-125b-5p, miR-451a, and miR-605-5p were differentially expressed in the EVs of YOAD patients when compared with HC. In LOAD patients, miR-125b-5p, miR-451a, and miR-605-5p were similarly altered in expression, but miR-16-5p showed similar expression to HC.[42]
miR-27a-3p,
miR-30a-5p,
miR-34c,
piR_019324,
piR_019949,
piR_020364
ADCSFAD patients (N = 42),
MCI patients (N = 17),
psychiatric and neurological controls (N = 82)
n.m.3500× g (10′ at 4 °C) + 2X 4500× g (10′ at 4 °C) + 10,000× g (30′ at 4 °C) +
ultracentrifugation
A combined signature consisting of three miRNAs and three piRNAs were suitable to detect AD with an AUC of 0.83. The piRNA signature could predict the conversion of MCI patients to AD with an AUC of 0.86. When combining the smallRNA signature with pTau and Aβ 42/40 ratio the AUC reaches 0.98.[61]
miR-23a-3p,
miR-223-3p,
miR-190a-5p,
miR-100-3p
ADPlasmaAD patients (N = 40),
HC (N = 40)
AD—25♀ + 15♂
(73 yrs)
HC—18♀ + 22♂
(67 yrs)
3000× g (15′) + Thrombin + 10,000 rpm (5′) + ExoQuick® (System Biosciences)
+ L1CAM IP
MiR-23a-3p, miR-223- 3p and miR-190a-5p levels in NDEVs from AD patients were significantly upregulated as compared with HCs, whereas miR-100-3p levels were significantly downregulated.[93]
miR-204-5p,
miR-632
FTDCSFGeNFI cohort:
GRN, C9orf72 and MAPT mutation carriers (N = 38),
Non-mutation carriers (N = 11);
Sporadic disease cohort:
FTD patients (N = 11),
PPA patients (N = 6),
AD patients (N = 13),
HC (N = 10)
Mutation carriers—25♀ + 13♂ (54 yrs)
Non-mutation carriers—6♀ + 5♂ (47 yrs)
FTD—2♀ + 9♂ (67 yrs)
PPA—2♀ + 4♂ (66yrs)
AD—5♀ + 8♂ (63 yrs)
HC—5♀ + 5♂ (69 yrs)
10,000× g (5′) +
miRCURYTM Exosome Isolation Kit
In the GeNFI cohort, miR-204-5p and miR-632 were significantly decreased in symptomatic compared with presymptomatic mutation carriers, with an AUC of 0.89 and 0.81, respectively,
and 0.93 when combined. In sporadic FTD, only miR-632 was significantly decreased compared with AD and HC (AUC = 0.90).
[47]
miR-233DementiaSerumDementia patients:
First clinic visit AD (ADfirst) (N = 11),
Treatment-receiving AD (ADcare) (N = 11),
VD (N = 10);
HC (N = 16)
ADfirst—5♀ + 6♂ (76 yrs)
ADcare—4♀ + 7♂ (79 yrs)
VD—4♀ + 6♂ (82 yrs)
HC—8♀ + 8♂ (80 yrs)
3000× g (15′ at 4 °C) + ExoQuick® The median levels of EVs miR-223 was significantly decreased in dementia patients, when comparing with HC (AUC = 0.875).[51]
miR-132-3p,
miR-212
AD and MCIPlasmaAD patients (N = 16),
AD-MCI patients (N = 16),
HC (N = 31)
n.m.Thrombin + 6000× g (20′ at 4 °C) + ExoQuick® + L1CAM IPMeasurement of miR-132-3p in NDEVs showed good sensitivity and specificity to diagnose AD, but did not effectively separate individuals with AD-MCI from HC. MiR-212 was also decreased in NDEVs from AD patients compared to HC.[55]
let-7e-5p,
miR- 125a-5p,
miR-23a-3p,
miR-375,
miR-1468-5p,
miR-204-5p,
miR-369-5p,
miR-423-5p
AD and PDPlasmaAD patients (N = 5),
PD patients (N = 7),
HC (N = 34)
AD—18♀ + 22♂
(67 yrs)
PD—6♀ + 1♂ (62 yrs)
HC—14♀ + 20♂
(33 yrs)
exoRNeasy Serum/Plasma Maxi Kit (QIAGEN) and
8000× g (5′) + ExoQuick®
Compared to the HC, eight miRNAs were found to be significantly elevated/declined in AD and PD samples, of which fiour miRNAs were newly identified.[87]
miR-1246,
miR-127-3p,
miR-19b-3p,
miR-134-5p,
miR-370- 3p,
miR-375,
miR-379-5p,
miR-382-5p,
miR-432-5p,
miR-485-5p,
miR-493-3p
MSSerumRRMS patients (N = 29)17♀ + 12♂ (34 yrs)SEC (qEV iZON Science) Several combinations of two or three miRNAs were able to distinguish active from quiescent disease with greater than 90% accuracy. Additional miRNAs associated with stable remission, and a positive response to fingolimod in patients with active disease prior to treatment.[79]
miR-9-5p,
miR-15a-5p,
miR-183-5p,
miR- 193a-5p,
miR-338-3p,
miR-1246
ALSPlasmaALS patients (N = 14),
HC (N = 8)
ALS—8♀ + 6♂ (62 yrs)
HC—n.m.
Vn96 peptide methodMiRNAs with relevance to ALS were found to be deregulated, including miR-9-5p, miR-183-5p, miR-338-3p and miR-1246. MiR-15a-5p and miR-193a-5p were identified for their di- agnostic potential of ALS and association with disability progression, respectively.[70]
miR-146a-5p,
miR-199a-3p,
miR-4454,
miR-10b-5p,
miR-29b-3p,
miR-151a-3p,
miR-151a-5p,
miR-199a-5p
ALS/MNDPlasmaALS/MND patients (N = 10 + 10),
HC (N = 10 + 10)
n.m.Thrombin + ExoQuick® + L1CAM IPFive upregulated and three downregulated miRNA sequences significantly distinguished ALS/MND patients from HC in two independent patient cohorts.[75]
miR-203b-5p,
miR-203a-3p,
miR-206,
miR- 185-5p
TBIPlasmaTBI patients (N = 16),
HC (N = 20)
n.m.Track Etched Magnetic Nanopore (TENPO) sorting for GluR2A panel of four miRNAs significantly discriminated TBI patients vs. HC.[63]
miR-139-5p,
miR-18a-5p,
miR-103a-3p
TBIPlasma1-2 TBIs patients (N = 73),
rTBI patients (N = 45),
HC (N = 35)
1-2 TBI—7♀ + 66♂ (39 yrs)
rTBI—9♀ + 36♂
(41 yrs)
HC—4♀ + 31♂
(42 yrs)
3000 rpm (5′) +
exoRNeasy Serum/Plasma Kit
MiR-139-5p and miR-18a-5p, were significantly differentially expressed in the rTBI and 1-2 TBI groups. TBI history and neurobehavioral symptom survey scores negatively correlated with miR-103a-3p expression.[78]
miR-9,
miR-124
AISSerumAIS patients (N = 65),
HC (N = 66)
AIS—25♀ + 40♂
(64 yrs)
HC—30♀ + 36♂
(60 yrs)
21,000× g (15′ at 4 °C) + ExoQuick®miR-9 and miR-124 were significantly higher in AIS patients vs. HC (AUCs of 0.8026 and 0.6976, respectively).[23]
miR-21-5p,
miR-30a-5p
ISPlasmaHIS patients (N = 15),
AIS patients days 1-3 (N = 33),
AIS patients days 3-7 (N = 32),
SIS patients (N = 31),
RIS patients (N = 32)
HC (N = 24)
HIS—5♀ + 10♂ (58 yrs)
AIS days 1-3—9♀ + 24♂ (58 yrs)
AIS days 3-7—13♀ + 19♂ (58 yrs)
SIS—9♀ + 22♂ (62 yrs)
RIS—4♀ + 28♂ (62 yrs)
HC—6♀ + 18♂ (57 yrs)
16,000× g (10′ at 4 °C) + exoRNeasy Serum/Plasma KitMiR- 21-5p and miRNA-30a-5p in combination are promising biomarkers for diagnosing IS and distinguishing among HIS, SIS, and RIS, especially miRNA-30a-5p for the diagnosis of the HIS phase.[50]
miR-122-3p,
miR-200a-5p
NMOSDSerumNMOSD in relapsing patients (N = 16),
NMOSD in remission patients (N = 15),
HC (N = 14)
NMOSD relapsing—14♀ + 2♂ (37 yrs)
NMOSD remission—13♀ + 2♂ (39 yrs)
HC—12♀ + 2♂ (35 yrs)
RiboTM Exosome Isolation Reagent (RiboBio)MiR-122-3p and miR-200a-5p could distinguish NMOSD status, and were significantly upregulated in the serum EVs of relapsing NMOSD compared with that in remitting NMOSD. The two miRNAs had positive correlations with disease severity in NMOSD patients.[76]
miR-3613-5p,
miR-4668-5p,
miR-8071,
miR-197-5p,
miR-4322,
miR-6781-5p
mTLE-HSPlasmamTLE-HS patients (N = 40),
HC (N = 40)
mTLE-HS—15♀ + 25♂ (27 yrs)
HC—n.m.
2000× g (20′) + 10,000× g (20′) + ExoQuick®Among six candidate microRNAs, miR-8071 had the best diagnostic value for mTLE-HS with 83.33% sensitivity and 96.67% specificity, and was associated with seizure severity.[32]
miR-206,
miR619-5p,
miR-133a-3p, miR-143-3p, miR-144-5p, miR-499a-5p, miR-3614-5p, miR-941, miR-30c-5p, miR-339-5p, miR-30b-5p, miR-6515-5p
SCZSerumSCZ patients (N = 100),
HC (N = 100)
SCZ—50♀ + 50♂
(30 yrs)
HC—42♀ + 58♂
(29 yrs)
SEC (qEV iZON Science) MiR-206 was the most upregulated miRNA in the EVs of SCZ patients. A signature of 11 miRNAs were identified in EVs from SCZ patients and were used to classify samples from SCZ and HC subjects with high accuracy.[58]
miR-203a-3pPTSDPlasmaDiscovery set:
PTSD patients (N = 12),
HC (N = 12);
validation set:
PTSD patients (N = 10),
HC (N = 10)
Discovery set:
PTSD—12♂ (31 yrs)
HC—12♂ (34 yrs)
validation set:
PTSD—10♂ (31 yrs)
HC—10♂ (31 yrs)
10,000× g (10′) + SEC (iZON Science)The concentration changes of miR-203a-3p in EV and miR-339-5p in EV-depleted plasma were confirmed two independent cohort veterans with PTSD.[65]
RNU6-1,
miR-320,
miR-574
GMSerumTraining set:
GM patients (N = 25),
HC (N = 25);
validation set:
GM patients (N = 50),
HC (N = 30)
Training set:
GM—11♀ + 14♂
(60 yrs)
HC—11♀ + 14♂
(60 yrs)
validation set:
GM—20♀ + 30♂
(61 yrs)
HC—16♀ + 14♂
(54 yrs)
ExoQuick®The expression levels of the sncRNA RNU6-1, miR-320 and miR-574-3p were significantly associated with a GM diagnosis. RNU6-1 was consistently an independent predictor of a GBM diagnosis.[17]
miR-21,
miR-27b,
miR-130b,
miR-193b,
miR-218,
miR-331,
miR-374a,
miR-520f,
miR-548c
GMCSFDiscovery cohort 1 patients (N = 24) and HC (N = 15);
discovery cohort 2 patients (N = 40) and HC (N = 27);
discovery cohort 3 patients (N = 13) and HC (N = 19);
validation cohort 4 patients (N = 10) and HC (N = 12);
validation cohort 5 patients (N = 18) and HC (N = 20)
Discovery cohort 1—17♀ + 22♂ (61 yrs)
discovery cohort 2—32♀ + 35♂ (59 yrs)
discovery cohort 3—13♀ + 19♂ (57 yrs)
validation cohort 4—5♀ + 17♂ (54 yrs)
validation cohort 5—25♀ + 15♂ (58 yrs)
2000× g (20′) +
ultracentrifugation
Comparison of miRNA profiles between GM patients and HC yielded a tumor “signature” consisting of nine miRNAs, which correlated with GM tumor volume.[27]
HOTAIRGMSerumGM patients (N = 43),
HC (N = 40)
n.m.Total Exosome Isolation Kit
(InvitrogenTM)
HOTAIR was present in whole serum and purified EVs but not in serum supernatant depleted of EVs, in GM patients.[49]
RNU6-1GMSerumGM patients (N = 18), subacute stroke patients (N = 30), acute/subacute hemorrhage patients (N = 30),
MS patients (N = 18),
brain metastases patients (N = 21), PCNSL patients (N = 12),
HC (n = 30),
GM—8♀ + 10♂ (63 yrs)
stroke—11♀ + 19♂ (71 yrs)
hemorrhage—9♀ + 21♂ (66 yrs)
MS—13♀ + 6♂ (41 yrs)
PCNSL—4♀ + 8♂ (66 yrs)
metastases—10♀ + 11♂ (60 yrs)
HC—17♀ + 13♂
(47 yrs)
ExoQuick®RNU6-1 expression was significantly higher in GM patients vs. HC, and also when comparing with patients with non-neoplastic lesions. No significant differences were found between GM patients and brain metastases.[91]
miR-21GliomaCSFGlioma patients (N = 70),
non-glioma controls (N = 25)
Glioma—28♀ + 42♂ (50 yrs)
non-glioma—7♀ + 18♂ (54 yrs)
2000× g (30′) + 12,000× g (25′) +
ultracentrifugation
MiR-21 levels in CSF-EVs of glioma patients were found significantly higher than in non-glioma controls; whereas no difference was detected in serum-derived EVs. The CSF-EVs miR-21 levels correlated with tumor spinal/ventricle metastasis and the recurrence with anatomical site preference.[20]
miR-4443,
miR-422a,
miR-494-3p,
miR-502-5p,
miR-520f-3p,
miR-549a
GliomaSerumGlioma patients (N = 28),
HC (N = 8)
Glioma—13♀ + 15♂ (49 yrs)
HC—n.m.
3 × 3500 rpm (20′) +
ExoQuick®
Six overexpressed miRNAs were found on EVs from glioma patients vs. HC. MiR-549a and miR-502-5p expression predicted prognosis in glioma patients.[33]
miR-301aGliomaSerumGlioma patients (N = 60),
HC (N = 43)
Glioma—33♀ + 27♂ (n.m.)
HC—n.m.
3000× g (15′ at 4 °C) + ExoQuick®MiR-301a levels on EVs were upregulated in glioma patients compared to HC, and correlated with ascending pathological grades. MiR-301a levels were significantly reduced after surgical resection of primary tumors and increased again during GM recurrence, and were independently associated with overall survival.[40]
miR-21,
miR-222,
miR-124-3p
GliomaSerumGlioma patients (N = 100),
brain non-glial metastases patients (N = 11),
HC (N = 30)
Glioma—40♀ + 60♂ (n.m.)
non-glial metastases—40♀ + 60♂ (n.m.)
HC—8♀ + 3♂ (41 yrs)
3000× g (15′) + ExoQuick®The expression levels of miR-21, miR-222 and miR-124-3p in EVs of patients with high grade gliomas were significantly higher than those of low grade gliomas and HC, and were decreased in samples obtained after surgery.[46]
miR-454-3pGliomaSerumGlioma patients (N = 24),
HC (N = 24)
n.m.RiboTM Exosome Isolation ReagentMiR-454-3p was significantly downregulated in tumor tissues, while it was upregulated in EVs from the same patients with glioma, corresponding to an AUC of 0.8663. MiR-454-3p expression was lower in the post-operative samples. High miR- 454-3p expression in EVs or low expression in tissues was associated with poor prognosis[71]
miR-210,
miR-5194,
miR-449
GliomaPlasmaGM patients (N = 25), LGA patients (N = 25),
Head trauma patients (N = 15)
GM—6♀ + 19♂ (n.m.)
LGA—10♀ + 15♂ (n.m.)
trauma—6♀ + 9♂ (n.m.)
n.m.MiR-210 was upregulated in GM and LGA, whereas miR-185, miR-5194, and miR-449 were downregulated in GM and LGA compared to trauma patients. MiR-5194 and miR-449 were significantly decreased in GM patients compared with LGA.[94]
Abbreviations: AD—Alzheimer’s disease; AIS—acute phase ischemic stroke; ALS—amyotrophic lateral sclerosis; CSF—cerebrospinal fluid; EV—extracellular vesicles; FTD—frontotemporal dementia; GM—glioblastoma multiforme; HC—healthy controls; HIS—hyperacute phase Ischemic stroke; IS—ischemic stroke; LGA—low-grade astrocytoma; MCI—mild cognitive impairment; miR—microRNA; MND—motor neuron disease; MS—multiple sclerosis; mTLE-HS—mesial temporal lobe epilepsy with hippocampal sclerosis; NDEV—neuron-derived extracellular vesicles; n.m.—not mentioned; NMOSD—Neuromyelitis optica spectrum disorders; PCNSL—primary central nervous system lymphoma; PD—Parkinson’s disease; PPA—primary progressive aphasia; PTSD—post-traumatic stress disorder; RIS—recovery phase ischemic stroke; RRMS—relapsing-remitting multiple sclerosis; rTBI—repetitive traumatic brain injury; SCZ—schizophrenia; SEC—size exclusion chromatography; SIS—subacute phase ischemic stroke; TBI—traumatic brain injury; VD—vascular dementia; yrs—years.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Monteiro-Reis, S.; Carvalho-Maia, C.; Bart, G.; Vainio, S.J.; Pedro, J.; Silva, E.R.; Sales, G.; Henrique, R.; Jerónimo, C. Secreted Extracellular Vesicle Molecular Cargo as a Novel Liquid Biopsy Diagnostics of Central Nervous System Diseases. Int. J. Mol. Sci. 2021, 22, 3267. https://doi.org/10.3390/ijms22063267

AMA Style

Monteiro-Reis S, Carvalho-Maia C, Bart G, Vainio SJ, Pedro J, Silva ER, Sales G, Henrique R, Jerónimo C. Secreted Extracellular Vesicle Molecular Cargo as a Novel Liquid Biopsy Diagnostics of Central Nervous System Diseases. International Journal of Molecular Sciences. 2021; 22(6):3267. https://doi.org/10.3390/ijms22063267

Chicago/Turabian Style

Monteiro-Reis, Sara, Carina Carvalho-Maia, Genevieve Bart, Seppo J. Vainio, Juliana Pedro, Eunice R. Silva, Goreti Sales, Rui Henrique, and Carmen Jerónimo. 2021. "Secreted Extracellular Vesicle Molecular Cargo as a Novel Liquid Biopsy Diagnostics of Central Nervous System Diseases" International Journal of Molecular Sciences 22, no. 6: 3267. https://doi.org/10.3390/ijms22063267

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop