Review
Promise of extracellular vesicles for diagnosis and treatment of epilepsy

https://doi.org/10.1016/j.yebeh.2019.106499Get rights and content

Highlights

  • Evaluation of brain-derived EVs in the blood may help in identifying biomarkers for distinct types of epilepsies.

  • Investigation of brain-derived EVs in the blood could help in identifying miRNAs involved in epileptogenesis.

  • EVs released from mesenchymal stem cells have therapeutic properties.

  • Stem cell-derived EV therapy appears attractive for counteracting epileptogenic changes after SE.

  • EVs can be engineered to deliver specific miRNAs, proteins, or antiepileptic drugs to the brain.

Abstract

Extracellular vesicles (EVs) released from cells play vital roles in intercellular communication. Moreover, EVs released from stem cells have therapeutic properties. This review confers the potential of brain-derived EVs in the cerebrospinal fluid (CSF) and the serum as sources of epilepsy-related biomarkers, and the promise of mesenchymal stem cell (MSC)-derived EVs for easing status epilepticus (SE)-induced adverse changes in the brain. Extracellular vesicles shed from neurons and glia in the brain can also be found in the circulating blood as EVs cross the blood–brain barrier (BBB). Evaluation of neuron and/or glia-derived EVs in the blood of patients who have epilepsy could help in identifying specific biomarkers for distinct types of epilepsies. Such a liquid biopsy approach is also amenable for repeated analysis in clinical trials for comprehending treatment efficacy, disease progression, and mechanisms of therapeutic interventions. Extracellular vesicle biomarker studies in animal prototypes of epilepsy, in addition, could help in identifying specific micro ribonucleic acid (miRNAs) contributing to epileptogenesis, seizures, or cognitive dysfunction in different types of epilepsy. Furthermore, intranasal (IN) administration of MSC-derived EVs after SE has shown efficacy for restraining SE-induced neuroinflammation, aberrant neurogenesis, and cognitive dysfunction in an animal prototype. Clinical translation of EV therapy as an adjunct to antiepileptic drugs appears attractive to counteract the progression of SE-induced epileptogenic changes, as the risk for thrombosis or tumor is minimal with nanosized EVs. Also, EVs can be engineered to deliver specific miRNAs, proteins, or antiepileptic drugs to the brain since they incorporate into neurons and glia throughout the brain after IN administration.

This article is part of the Special Issue “NEWroscience 2018"

Introduction

A variety of vesicles released by cells into the extracellular space are generically referred to as extracellular vesicles (EVs) [1]. Extracellular vesicles, found in all body fluids, are delimited by a phospholipid bilayer and contain nucleic acids and proteins from donor cells [2]. Extracellular vesicles partake in intercellular communication and likely also influence many cellular processes. Studies characterizing the composition of EVs in various body fluids have suggested that an analysis of EVs from the serum or the cerebrospinal fluid (CSF) could provide information on specific biomarkers in various neurodegenerative diseases. Investigation of EVs isolated from the serum or CSF has advantages because such characterization allows the analysis of the composition of EVs secreted from specific brain cells (e.g., EVs derived from neurons vis-à-vis astrocytes). Also, the configuration of EVs is likely more stable than soluble biomarkers found in the CSF or the serum.

Extracellular vesicles are broadly classified as microvesicles (MVs) or exosomes (EXs) based on their intracellular origin. Microvesicles bud off from the plasma membrane and are rich in negatively charged phospholipids such as phosphatidylserine at their surface [2] (Fig. 1). The size of MVs can vary from 100 to 1000 nm [3]. The composition of MVs depends on the cell type from which they are generated and the type of stimulation causing their creation. However, proteins specific to MVs are yet to be identified [2]. In contrast, EXs originate in endosomes as intraluminal vesicles through inward invagination of the endosomal membrane, which eventually results in the formation of multivesicular bodies (MVBs) containing a constellation of EXs (Fig. 1). While some MVBs fuse with lysosomes to degrade their cargo, others fuse with the plasma membrane to secrete EXs into the extracellular space [4] (Fig. 1). The creation of EXs, particularly the formation of intraluminal vesicles, requires the involvement of intricate protein machinery termed the endosomal sorting complex required for transport (ESCRT). Nonetheless, the formation of EXs can also occur in an ESCRT-independent manner, which involves the participation of syndecan/syntenin/ALG-2-interacting protein X (ALIX) pathway [5]. Besides, CD9- and CD63-dependent pathways have also been implicated in EX formation [2], [6], [7]. The EXs exhibit well-demarcated round morphology, and their size can vary from 30 to 100 nm, depending on the type of cells from which they are released [8], [9], [10], [11]. While the kinetics, the composition, and the biological properties of EXs vary significantly in health and disease conditions [12], EXs from various sources have been isolated, characterized, and customized for specific applications. Analyses of the composition of EXs derived from multiple sources have implied that EXs are rich in proteins, lipids, and nucleic acids derived from their parental cells.

The details on the composition of proteins, lipids, and RNA in EVs or EXs can be seen in three different web-based resources, either at http://www.exocarta.org/ or Vesiclepedia or EVpedia. These databases suggest frequently observed protein components in EVs and EXs, which include annexins, cytoskeletal proteins, heat shock proteins, integrins, metabolic enzymes, ribosomal proteins, tetraspanins, and vesicle trafficking-related proteins [13]. The major lipid components of EXs derived from various cells are recently reviewed [14], which includes cholesterol, sphingomyelin, phosphatidylcholine, phosphatidylserine, phosphatidylethanolamine, phosphatidylethanolamine ethers, diacylglycerol, phosphatidylcholine ethers, hexosyl ceramide, ceramides, phosphatidylglycerol, phosphatidic acid, phosphatidylinositol, lactosylceramide, lysophosphatidylinositol, lysophosphatidylethanolamine, cholesteryl ester, globotriaosylceramide, etc. [14]. The nucleic acids in EXs comprise mRNAs, microRNAs (miRNAs), and other noncoding RNAs. While the composition of EXs varies depending on the type of cell from which EXs are released, the exact mechanism by which a variety of molecules are loaded into EXs is still unknown. Interestingly, the protein composition of EXs is more likely to be different from their mother cells than the protein composition in MVs [15]. Moreover, EXs tend to be enriched with proteins of extracellular matrix, heparin-binding receptors, and immune response and cell adhesion functions. Conversely, MVs are enriched with proteins from the endoplasmic reticulum, proteasome, and mitochondria [2]. Following their release by donor cells, EXs can enter recipient cells, influence their function by activating intracellular signaling pathways through the transfer of their cargo. The uptake of EXs or its cargo by recipient cells can occur through several mechanisms. For instance, endocytosis of EXs depends on caveolae in epithelial cells [16], clathrin in neurons [17], and cholesterol and lipid raft in endothelial cells [18]. The incorporation of EXs by microglia and macrophages involves macropinocytosis [19] or phagocytosis [20].

Since the size range of MVs and EXs overlaps, a rigorous characterization of EV properties is necessary for classifying them as either MVs or EXs. Initially, an evaluation of at least three specific markers was considered essential to classify EVs as EXs [21]. The markers include tetraspanins CD9, CD63, CD81, the endosomal proteins tumor susceptibility gene 101 (TSG101), and ALIX. Because some of these markers can also be found in MVs [22], recent guidelines from the International Society for Extracellular Vesicles (ISEV) suggest the use of operational terms unless the release of EXs by cells is caught in the act through live imaging techniques. The suggested operational terms are based on physical characteristics of EVs such as the size (small EVs, medium/large EVs), biochemical composition (e.g., CD63 +/CD81 +-/CD9 +- EVs), or cells of origin (e.g., neuron-derived EVs, astrocyte-derived EVs, or microglia-derived EVs) [1]. Therefore, we employed the phrase “EVs” for all EV or EX studies discussed in this review in the subsequent sections.

The goal of this article was to confer two specific issues about EVs. The first segment of the review deliberates the potential of EVs in body fluids, particularly in the serum and the CSF, as sources of specific epilepsy biomarkers. The second part of the review confers the promise of mesenchymal stem cell (MSC)-derived EVs for treating status epilepticus (SE)-induced brain dysfunction and chronic epilepsy.

Section snippets

Potential of EVs as biomarkers of epilepsy

Several disease-specific proteins and/or miRNAs are enriched in EVs shed from neurons or glia into the blood or the CSF. Therefore, evaluation of the composition of EVs released into the plasma or the CSF by neurons and glia in the central nervous system (CNS) can provide diagnostic and/or prognostic insights on brain function particularly in neurological disorders [2], [23]. Especially, neuron-, astrocyte-, oligodendrocyte- and microglia-derived EVs (NDEVs, ADEVs, ODEVs, MDEVs) in the serum or

Therapeutic potential of mesenchymal stem cell-derived EVs for modulating brain dysfunction after SE or in chronic epilepsy

Multiple studies have demonstrated the beneficial effects of MSC-derived EVs, for treating neurological conditions such as stroke [43], ischemia [44], traumatic brain injury [10], [45], hypoxic-ischemia-induced perinatal brain injury [46], [47], preterm brain injury [48], multiple sclerosis [49], AD [50], and spinal cord injury [51]. Kim and colleagues investigated the efficacy of human MSC-derived EVs in a controlled cortical impact injury model [10]. Extracellular vesicles employed in this

Summary and future perspectives

Extracellular vesicles are enriched with miRNAs, in comparison with non-EV fractions in the CSF or plasma. Since many studies have confirmed the occurrence of higher miRNA levels in EVs, characterization of CNS-derived EVs such as NDEVs, ADEVs, ODEVs, or MDEVs from the CSF and/or plasma for miRNA expression appears highly useful for biomarker discovery in various types of epilepsy. Notably, analysis of NDEVs in the serum has received considerable attention for evaluating brain biomarkers in

Declaration of Competing Interests

None.

Acknowledgments

Authors are supported by grants from the National Institute of Neurological Disorders and Stroke (1R01NS106907-01 to DJP and AKS), the Department of Defense (W81XWH-14-1-0558 to A.K.S.), the State of Texas (Emerging Technology Fund to A.K.S.), and Science and Engineering Research Board (SERB -EMR/2017/005213 to DU).

References (76)

  • K. Drommelschmidt et al.

    Mesenchymal stem cell-derived extracellular vesicles ameliorate inflammation-induced preterm brain injury

    Brain Behav Immun

    (2017)
  • X. Zhou et al.

    Mesenchymal stem cell derived EVs mediate neuroprotection after spinal cord injury in rats via the microRNA-21-5p/FasL gene axis

    Biomed Pharmacother

    (2019)
  • J.A. Gorter et al.

    Status epilepticus, blood-brain barrier disruption, inflammation, and epileptogenesis

    Epilepsy Behav

    (2015)
  • Z.S. Costa-Ferro et al.

    Transplantation of bone marrow mononuclear cells decreases seizure incidence, mitigates neuronal loss and modulates pro-inflammatory cytokine production in epileptic rats

    Neurobiol Dis

    (2012)
  • A. Vezzani et al.

    The role of inflammation in epileptogenesis

    Neuropharmacology

    (2013)
  • A.K. Shetty

    Hippocampal injury-induced cognitive and mood dysfunction, altered neurogenesis, and epilepsy: can early neural stem cell grafting intervention provide protection?

    Epilepsy Behav

    (2014)
  • S.M. Ryan et al.

    Neuroinflammation negatively affects adult hippocampal neurogenesis and cognition: can exercise compensate?

    Neurosci Biobehav Rev

    (2016)
  • B. Hattiangady et al.

    Incorporation of embryonic CA3 cell grafts into the adult hippocampus at 4-months after injury: effects of combined neurotrophic supplementation and caspase inhibition

    Neurosci

    (2006)
  • M.S. Hester et al.

    Impact of rapamycin on status epilepticus induced hippocampal pathology and weight gain

    Exp Neurol

    (2016)
  • E.V. Batrakova et al.

    Using exosomes, naturally-equipped nanocarriers, for drug delivery

    J Control Release

    (2015)
  • A. Vogel et al.

    Neural stem cell derived extracellular vesicles: attributes and prospects for treating neurodegenerative disorders

    EBioMedicine.

    (2018)
  • C. Théry et al.

    Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines

    J Extracell Vesicles

    (2018)
  • M. Malloci et al.

    Extracellular vesicles: mechanisms in human health and disease

    Antioxid Redox Signal

    (2019)
  • S. Flamant et al.

    Extracellular vesicles and vascular injury: new insights for radiation exposure

    Radiat Res

    (2016)
  • G. Raposo et al.

    Extracellular vesicles: exosomes, microvesicles, and friends

    J Cell Biol

    (2013)
  • M.F. Baietti et al.

    Syndecansyntenin-ALIX regulates the biogenesis of exosomes

    Nat Cell Biol

    (2012)
  • S.I. Buschow et al.

    MHC II in dendritic cells is targeted to lysosomes or T cell-induced exosomes via distinct multivesicular body pathways

    Traffic

    (2009)
  • B.-T. Pan et al.

    Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes

    J Cell Biol

    (1985)
  • M. Ostrowski et al.

    Rab27a and Rab27b control different steps of the exosome secretion pathway

    Nat Cell Biol

    (2010)
  • D.K. Kim et al.

    Chromatographically isolated CD63 + CD81 + extracellular vesicles from mesenchymal stromal cells rescue cognitive impairments after TBI

    Proc Natl Acad Sci U S A

    (2016)
  • M. Durcin et al.

    Characterisation of adipocyte-derived extracellular vesicle subtypes identifies distinct protein and lipid signatures for large and small extracellular vesicles

    J Extracell Vesicles

    (2017)
  • J. De Toro et al.

    Emerging roles of exosomes in normal and pathological conditions: new insights for diagnosis and therapeutic applications

    Front Immunol

    (2015)
  • S. Mathivanan et al.

    ExoCarta: a compendium of exosomal proteins and RNA

    Proteomics

    (2009)
  • R.A. Haraszti et al.

    High-resolution proteomic and lipidomic analysis of exosomes and microvesicles from different cell sources

    J Extracell Vesicles

    (2016)
  • A. Nanbo et al.

    Exosomes derived from Epstein-Barr virus-infected cells are internalized via caveola-dependent endocytosis and promote phenotypic modulation in target cells

    J Virol

    (2013)
  • C. Frühbeis et al.

    Neurotransmitter-triggered transfer of exosomes mediates oligodendrocyte-neuron communication

    PLoS Biol

    (2013)
  • D. Fitzner et al.

    Selective transfer of exosomes from oligodendrocytes to microglia by macropinocytosis

    J Cell Sci

    (2011)
  • D. Feng et al.

    Cellular internalization of exosomes occurs through phagocytosis

    Traffic

    (2010)
  • Cited by (23)

    View all citing articles on Scopus
    View full text