Summary

Enrichment of Astrocyte-Derived Extracellular Vesicles from Human Plasma

Published: August 03, 2022
doi:

Summary

This protocol describes the enrichment of astrocyte-derived extracellular vesicles (ADEVs) from human plasma. It is based on the separation of EVs by polymer precipitation, followed by ACSA-1 based immunocapture of ADEVs. Analysis of ADEVs may offer clues to study changes in inflammatory pathways of living patients, non-invasively by liquid biopsy.

Abstract

Extracellular vesicles (EVs) are biological nanoparticles secreted by all cells for cellular communication and waste elimination. They participate in a vast range of functions by acting on and transferring their cargos to other cells in physiological and pathological conditions. Given their presence in biofluids, EVs represent an excellent resource for studying disease processes and can be considered a liquid biopsy for biomarker discovery. An attractive aspect of EV analysis is that they can be selected based on markers of their cell of origin, thus reflecting the environment of a specific tissue in their cargo. However, one of the major handicaps related to EV isolation methods is the lack of methodological consensuses and standardized protocols. Astrocytes are glial cells with essential roles in the brain. In neurodegenerative diseases, astrocyte reactivity may lead to altered EV cargo and aberrant cellular communication, facilitating/enhancing disease progression. Thus, analysis of astrocyte EVs may lead to the discovery of biomarkers and potential disease targets. This protocol describes a 2-step method of enrichment of astrocyte-derived EVs (ADEVs) from human plasma. First, EVs are enriched from defibrinated plasma via polymer-based precipitation. This is followed by enrichment of ADEVs through ACSA-1-based immunocapture with magnetic micro-beads, where resuspended EVs are loaded onto a column placed in a magnetic field. Magnetically labeled ACSA-1+ EVs are retained within the column, while other EVs flow through. Once the column is removed from the magnet, ADEVs are eluted and are ready for storage and analysis. To validate the enrichment of astrocyte markers, glial fibrillary acidic protein (GFAP), or other specific astrocytic markers of intracellular origin, can be measured in the eluate and compared with the flow-through. This protocol proposes an easy, time-efficient method to enrich ADEVs from plasma that can be used as a platform to examine astrocyte-relevant markers.

Introduction

Extracellular vesicles (EVs) are a heterogeneous group of membranous nanoparticles secreted by all types of cells, carrying proteins, lipids, and nucleic acids1. Microvesicles (100-1000 nm), exosomes (30-100 nm), and apoptotic bodies (1000-5000 nm) constitute the main EV types, as distinguished by their site of origin2,3. EVs regulate important physiological processes, such as antigen presentation and immune responses4, receptor recycling, metabolite elimination5, and cellular communication6. The regulation of these processes may occur by direct binding between proteins enriched in the EV cell membrane and targets in recipient cells, and/or through the internalization and release of their cargo in the cytoplasm of the recipient cell7. While EVs perform essential cellular functions, they have gained increasing interest from a pathological perspective in the fields of cancer and neurology. Indeed, several studies have shown EVs can help promote tumor cell migration8,9 or seed toxic protein aggregates in neurodegenerative diseases, such as Alzheimer's disease10,11.

EVs can be selected and enriched from biofluids based on cell surface markers related to their cell of origin, thus reflecting the environment of a specific tissue in their cargo12,13,14,15,16,17,18,19,20. In addition, given their presence in blood, cerebrospinal fluid (CSF), saliva, urine, and breast milk, EVs represent an excellent, non-invasive tool for diagnosis, and can be considered a liquid biopsy for biomarker discovery. This is of special interest in neurology, given the difficulties of studying brain analytes in accessible fluids other than CSF.

Astrocytes have gained rising interest, as they are at the intersection of neuro-vascular communication21. Under physiological conditions, they are responsible for the preservation of the blood-brain barrier, the recycling of neurotransmitters, the supply of nutrients and growth factors to neurons and other glial cells22,23,24 as well as neuro-immune defense, given their metabolic plasticity from pro-inflammatory to anti-inflammatory states and vice versa25,26,27. An important mechanism by which astrocytes accomplish their regulatory functions is by communication through EVs28,29. Reactive astrocytosis is a key hallmark of several neurodegenerative diseases, such as Alzheimer's disease,30 multiple system atrophy (MSA), progressive supranuclear palsy (PSP)31, and amyotrophic lateral sclerosis (ALS)32. Astrocyte reactivity may lead to altered EV cargo, release of inflammatory mediators, and aberrant cellular communication, thus facilitating the spread of pathology and leading to neurodegeneration10,11. Therefore, studying astrocyte derived EVs (ADEVs) and changes in their cargo is an attractive resource to examine neurodegenerative processes in a non-invasive manner.

Currently, several methodologies exist for the isolation of EVs, each with its corresponding advantages and disadvantages33. It is essential to consider which method is more suitable for a specific use, depending on the final application of interest. In the neurology field, and more specifically, in astrocyte studies, polymer-based precipitation followed by immunocapture has been the predominantly used method12,18,19,20,34. However, even when applying the same approach, there remains heterogeneity between studies in the different steps applied for EV isolation. Therefore, there is a need for a clear, step-by-step standardized methodology to facilitate astrocyte EV studies and study reproducibility. Polymer-based precipitation facilitates biomarker screening given that it is a fast, simple procedure that does not require complex equipment, leading to a high yield of EVs without affecting their biological activity35.

The present protocol describes a detailed, simple, two-step method for the enrichment of ADEVs from human plasma. It is based on a polymer-based precipitation of the total EV fraction, followed by an immunocapture of astrocyte EVs. Given the important functions of astrocytes, analysis of ADEVs may shed light for the discovery of biomarkers and brain inflammatory pathways that can be studied in a non-invasive manner.

Protocol

The research described in this protocol has been conducted with human plasma samples from healthy adult donors of both sexes (age range 65.9-81.3 years, 45.5% females), from the Sant Pau Initiative on Neurodegeneration (SPIN) cohort, Barcelona, Spain36. Participants gave informed consent. The study has been conducted following the international ethical guidelines for medical research contained in the declaration of Helsinki and the Spanish law. The Sant Pau Research Ethics Committee (CEIC) reviewe…

Representative Results

The isolation of ADEVs from plasma collected from healthy donors was successfully accomplished. A polymer-based precipitation method was employed to obtain the total EV fraction, followed by an immunocapture with magnetic microbeads to obtain ADEVs. Western blot analysis of the total EV fraction prior to the immuno-capture step indicated the lack of calnexin (cellular contamination marker) and the presence of Alix and the transmembrane protein CD9 in the EV preparations (F…

Discussion

EVs have gained strong interest in biomedical research due to their diagnostic and therapeutic potential. Currently, one of the major handicaps related to EV isolation methods is the lack of methodological consensus and standardized protocols. This study provides a detailed protocol for the enrichment of astrocyte EVs from human plasma via polymer-based precipitation and GLAST immunocapture.

Different methodologies exist for the isolation of EVs from body fluids, each with their own a…

Disclosures

The authors have nothing to disclose.

Acknowledgements

The authors would like to acknowledge the help of Soraya Torres, Shaimaa El Bounasri El Bennadi, and Oriol Sanchez Lopez for sample handling and preparation. We would also like to acknowledge the collaboration of José Amable Bernabé, from the ICTS "NANBIOSIS", unit 6 (Unit of CIBER in Bioingineering, Biomaterials & Nanomedicine) of the Barcelona Materials Science Institute, Marti de Cabo Jaume from the Electron Microscopy Unit at Universitat Autonoma de Barcelona, Dr. Marta Soler Castany and Lia Ros Blanco from the Flow Cytometry Platform at Sant Pau Biomedical Research Institute (IIB-Sant Pau), as well as Dr. Joan Carles Escolà-Gil from the Pathophysiology of lipid-related diseases group at IIB-Sant Pau for help with the NTA, cryo-EM, Luminex, and ApoB determinations, respectively.

The authors acknowledge financial support from the Jérôme Lejeune Foundation (Project #1941 and #1913 to MFI and MCI), Instituto de Salud Carlos III (PI20/01473 to JF, PI20/01330 to AL, PI18/00435 to DA, and INT19/00016 to DA), the National Institute of Health (1R01AG056850-01A1, R21AG056974, and R01AG061566 to JF), the Alzheimer's Association and Global Brain Health Institute (GBHI_ALZ-18-543740 to MCI), the The Association for  Frontotemporal Degeneration (Clinical Research Postdoctoral Fellowship,  AFTD 2019–2021) to ODI, and the Societat Catalana de Neurologia (Premi Beca Fundació SCN 2020 to MCI). This work was also supported by the CIBERNED program (Program 1, Alzheimer Disease to AL and SIGNAL study. SS is a recipient of a Postdoctoral grant “Juan de la Cierva-Incorporación” (IJC2019-038962-I) by the Agencia Estatal de Investigación, Ministerio de Ciencia e Innovación (Gobierno de España).

Materials

Anti-Alix primary antibody for Western blotting EMD Millipore ABC40
µMACS Separator Miltenyi Biotec 130-042-602 The µMACS Separator is used in combination with µ Columns and MACS MicroBeads.
Anti-calnexin primary antibody for Western blotting Genetex GTX109669
Anti-CD9 primary antibody for Western blotting Cell Signaling 13174
Blocker BSA (10%) 200 mL Thermo Fisher 37525
Bransonic 1510E-MT Ultrasonic bath Branson
COBAS 6000 autoanalyzer Roche Diagnostics Analyzer for immunoturbidimetric determination of ApoB; commercial autoanalyzer
cOmplete Protease Inhibitor Cocktail (EDTA-free) Roche 11873580001
Digital Micrograph 1.8  micrograph software
Dulbecco's PBS Mg++, Ca++ free 500 mL Thermo Fisher 14190144
EveryBlot Blocking Buffer BioRad 12010020
Exoquick (exosome precipitation solution 5 mL) + Thrombin  System Bioscience EXOQ5TM-1 ExoQuick 20 mL can also be purchased (EXOQ20A-1)
Gatan 895 USC 4000  camera
GeneGnome XRQ chemiluminiscence imaging system Syngene
Human CD81 antigen (CD81) ELISA kit Cusabio CSB-EL004960HU
Human Programmed cell death 6-interacting protein (PDCD6IP) ELISA kit Cusabio CSB-EL017673HU
Immun-Blot PVDF Membrane BioRad 1620177
JEOL 2011 transmission electron microscope  JEOL LTD Equipped with a CCD Gatan 895 USC 4000 camera (Gatan 626, Gatan, Pleasanton, USA)
Lavender EDTA BD Vacutainer K2E tubes Becton dickinson 367525
Leica EM GP  Leica Microsystem commercial plunge freezer
Low binding microtubes 1,5 mL Deltalab 4092.3NS
MACS µ Columns with plungers  Miltenyi Biotec 130-110-905 µ Columns with plungers are especially designed for isolation of exosomes from body fluids
MACS Multistand Miltenyi Biotec 130-042-303
MAGPIX plate reader Luminex Corporation 80-073 Luminex's xMAP multiplexing unit (Luminex xPonent v 4.3 software)
MicroBead Kit100 μL Anti-GLAST (ACSA-1)-Biotin, human, mouse, rat – small size; 100 μL Anti-Biotin MicroBeads Miltenyi Biotec 130-095- 825
MILLIPLEX MAP Kit Human cytokine/Chemokine/Growth Factor Panel A magnetic bead panel EMD Millipore HCYTA-60K-25
M-PER Mammalian Protein Extraction Reagent 25 mL Thermo Fisher 78503 For certain applications like Western blot, more aggressive lysis buffers can be used (e.g. RIPA)
MultiSkan SkyHigh Microplate Spectrophotometer Thermofisher A51119500C
NanoSight NS300 Malvern Panalytical NTA; 3.4 version
Pierce Halt Protease and Phosphatase Inhibitor Cocktail Thermo Fisher 78441
Polypropylene syringe (G29) PeroxFarma 1mL syringe; 0.33x12mm-G29x1/2"
Secondary anti-rabbit antibody Thermo Fisher 10794347
Simoa GFAP Discovery Kit Quanterix 102336
Simoa, SR-X instrument Quanterix SR-X Ultra-Sensitive Biomarker Detection System; commercial biomarker detection technology
Specific Protein Test Apolipoprotein B – APOB (100 det) COBAS C/CI Roche Diagnostics 3032574122
SuperSignal West Femto Thermo Fisher 34095 Ultra-sensitive enhanced chemiluminescent (ECL) HRP substrate 
Trans-Blot Turbo Transfer System BioRad 1704150

References

  1. Pathan, M., et al. Vesiclepedia 2019: a compendium of RNA, proteins, lipids and metabolites in extracellular vesicles. Nucleic Acids Research. 47, 516-519 (2019).
  2. Raposo, G., Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. Journal of Cell Biology. 200 (4), 373-383 (2013).
  3. Gustafson, D., Veitch, S., Fish, J. E. Extracellular vesicles as protagonists of diabetic cardiovascular pathology. Frontiers in Cardiovascular Medicine. 4, 71 (2017).
  4. Raposo, G., et al. B lymphocytes secrete antigen-presenting vesicles. The Journal of Experimental Medicine. 183 (3), 1161-1172 (1996).
  5. Harding, C. V., Heuser, J. E., Stahl, P. D. Exosomes: Looking back three decades and into the future. Journal of Cell Biology. 200 (4), 367-371 (2013).
  6. Valadi, H., et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature Cell Biology. 9 (6), 654-659 (2007).
  7. Van Niel, G., D’Angelo, G., Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nature Reviews Molecular Cell Biology. 19 (4), 213-228 (2018).
  8. Hood, J. L., San Roman, S., Wickline, S. A. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. 암 연구학. 71 (11), 3792-3801 (2011).
  9. Rak, J. Microparticles in cancer. Seminars in Thrombosis and Hemostasis. 36 (8), 888-906 (2010).
  10. Ledreux, A., et al. Small neuron-derived extracellular vesicles from individuals with down syndrome propagate tau pathology in the wildtype mouse brain. Journal of Clinical Medicine. 10 (17), 3931 (2021).
  11. Winston, C. N., et al. Prediction of conversion from mild cognitive impairment to dementia with neuronally derived blood exosome protein profile. Alzheimer’s and Dementia: Diagnosis, Assessment and Disease Monitoring. 3, 63-72 (2016).
  12. Goetzl, E. J., Schwartz, J. B., Abner, E. L., Jicha, G. A., Kapogiannis, D. High complement levels in astrocyte-derived exosomes of Alzheimer’s disease. Annals of Neurology. 83 (3), 544-552 (2018).
  13. Fiandaca, M. S., 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 and Dementia. 11 (6), 600-607 (2015).
  14. Mustapic, M., et al. Plasma extracellular vesicles enriched for neuronal origin: A potential window into brain pathologic processes. Frontiers in Neuroscience. 11, 278 (2017).
  15. Hamlett, E. D., et al. Neuronal exosomes reveal Alzheimer’s disease biomarkers in Down syndrome. Alzheimer’s and Dementia. 13 (5), 541-549 (2017).
  16. Goetzl, E. J., et al. Abnormal levels of mitochondrial proteins in plasma neuronal extracellular vesicles in major depressive disorder. Molecular Psychiatry. 26 (12), 7355-7362 (2021).
  17. Goetzl, E. J., et al. Neuron-derived exosome proteins may contribute to progression from repetitive mild traumatic brain injuries to chronic traumatic encephalopathy. Frontiers in Neuroscience. 13, 452 (2019).
  18. Goetzl, E. J., et al. Cargo proteins of plasma astrocyte-derived exosomes in Alzheimer’s disease. FASEB Journal. 30 (11), 3853-3859 (2016).
  19. Goetzl, E. J., et al. Traumatic brain injury increases plasma astrocyte-derived exosome levels of neurotoxic complement proteins. FASEB Journal. 34 (2), 3359-3366 (2020).
  20. Nogueras-ortiz, C. J., et al. Astrocyte- and neuron-derived extracellular vesicles from Alzheimer’s disease patients effect complement-mediated neurotoxicity. Cells. 9 (7), 1618 (2020).
  21. Haydon, P. G., Carmignoto, G. Astrocyte control of synaptic transmission and neurovascular coupling. Physiological Reviews. 86 (3), 1009-1031 (2006).
  22. Abbott, N. J., Rönnbäck, L., Hansson, E. Astrocyte-endothelial interactions at the blood-brain barrier. Nature Reviews Neuroscience. 7 (1), 41-53 (2006).
  23. Rothstein, J., et al. Antisense knockout of glutamate transporters reveals a predominant role for astroglial glutamate transport in excitotoxicity and clearance of extracellular glutamate. Neuron. 16 (3), 675-686 (1996).
  24. Vasile, F., Dossi, E., Rouach, N. Human astrocytes: structure and functions in the healthy brain. Brain Structure and Function. 222 (5), 2017-2029 (2017).
  25. Balu, D. T., et al. Neurotoxic astrocytes express the D-serine synthesizing enzyme, serine racemase, in Alzheimer’s disease. Neurobiology of Disease. 130, 104511 (2019).
  26. Sofroniew, M. V. Astrocyte barriers to neurotoxic inflammation. Nature Reviews Neuroscience. 16 (5), 249-263 (2015).
  27. Yun, S. P., et al. Block of A1 astrocyte conversion by microglia is neuroprotective in models of Parkinson’s disease. Nature Medicine. 24 (7), 931-938 (2018).
  28. Bianco, F., et al. Acid sphingomyelinase activity triggers microparticle release from glial cells. EMBO Journal. 28 (8), 1043-1054 (2009).
  29. Datta Chaudhuri, A., et al. Stimulus-dependent modifications in astrocyte-derived extracellular vesicle cargo regulate neuronal excitability. Glia. 68 (1), 128-144 (2020).
  30. Serrano-Pozo, A., Gómez-Isla, T., Growdon, J. H., Frosch, M. P., Hyman, B. T. A phenotypic change but not proliferation underlies glial responses in Alzheimer disease. American Journal of Pathology. 182 (6), 2332-2344 (2013).
  31. Tong, J., et al. Low levels of astroglial markers in Parkinson’s Disease: relationship to α-synuclein accumulation. Neurobiology of Disease. 82, 243-253 (2015).
  32. Kamo, H., et al. A distinctive distribution of reactive astroglia in the precentral cortex in amyotrophic lateral sclerosis. Acta Neuropathologica. 74 (1), 33-38 (1987).
  33. Coumans, F. A. W., et al. Methodological guidelines to study extracellular vesicles. Circulation Research. 120 (10), 1632-1648 (2017).
  34. Winston, C. N., Goetzl, E. J., Schwartz, J. B., Elahi, F. M., Rissman, R. A. Complement protein levels in plasma astrocyte-derived exosomes are abnormal in conversion from mild cognitive impairment to Alzheimer’s disease dementia. Alzheimer’s and Dementia: Diagnosis, Assessment and Disease Monitoring. 11, 61-66 (2019).
  35. Niu, Z., et al. Polymer-based precipitation preserves biological activities of extracellular vesicles from an endometrial cell line. PLoS ONE. 12 (10), 1-21 (2017).
  36. Alcolea, D., et al. The Sant Pau Initiative on Neurodegeneration (SPIN) cohort: A data set for biomarker discovery and validation in neurodegenerative disorders. Alzheimer’s and Dementia: Translational Research and Clinical Interventions. 5, 597-609 (2019).
  37. O’Bryant, S. E., et al. Guidelines for the standardization of preanalytic variables for blood-based biomarker studies in Alzheimer’s disease research. Alzheimer’s and Dementia. 11 (5), 549-560 (2015).
  38. Kim, K., et al. Role of excitatory amino acid transporter-2 (EAAT2) and glutamate in neurodegeneration: Opportunities for developing novel therapeutics. Journal of Cellular Physiology. 226 (10), 2484-2493 (2011).
  39. Middeldorp, J., Hol, E. M. GFAP in health and disease. Progress in Neurobiology. 93 (3), 421-443 (2011).
  40. Mora, J., et al. Next generation ligand binding assays-review of emerging technologies’ capabilities to enhance throughput and multiplexing. AAPS Journal. 16 (6), 1175-1184 (2014).
  41. Chunyk, A. G., et al. A multi-site in-depth evaluation of the quanterix simoa from a user’s perspective. AAPS Journal. 20 (1), 1-12 (2018).
  42. Lötvall, J., et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: A position statement from the International Society for Extracellular Vesicles. Journal of Extracellular Vesicles. 3 (1), 26913 (2014).
  43. Hansen, E. O., et al. Millipore xMap® Luminex (HATMAG-68K): An accurate and cost-effective method for evaluating Alzheimer’s biomarkers in cerebrospinal fluid. Frontiers in Psychiatry. 12, 1-9 (2021).
  44. Kowal, J., et al. Proteomic comparison defines novel markers to characterize heterogenous populations of extracellular vesicle subtypes. Proceedings of the National Academy of Sciences. 113 (8), 968-977 (2016).
  45. Gauthier, S. A., et al. Enhanced exosome secretion in Down syndrome brain – a protective mechanism to alleviate neuronal endosomal abnormalities. Acta Neuropathologica Communications. 5 (1), 65 (2017).
  46. Cataldo, A. M., et al. Endocytic pathway abnormalities precede amyloid β deposition in sporadic alzheimer’s disease and down syndrome: Differential effects of APOE genotype and presenilin mutations. American Journal of Pathology. 157 (1), 277-286 (2000).
  47. Cataldo, A. M., et al. Down syndrome fibroblast model of Alzheimer-related endosome pathology: Accelerated endocytosis promotes late endocytic defects. American Journal of Pathology. 173 (2), 370-384 (2008).
  48. Théry, C., 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. Journal of Extracellular Vesicles. 7 (1), 1535750 (2018).
  49. Van Deun, J., et al. EV-TRACK: Transparent reporting and centralizing knowledge in extracellular vesicle research. Nature Methods. 14 (3), 228-232 (2017).
This article has been published
Video Coming Soon
Keep me updated:

.

Cite This Article
Valle-Tamayo, N., Pérez-González, R., Chiva-Blanch, G., Belbin, O., Serrano-Requena, S., Sirisi, S., Cervantes González, A., Giró, O., Sánchez-Aced, É., Dols-Icardo, O., Alcolea, D., Carmona-Iragui, M., Jimenez, A., Lleó, A., Fortea, J., Iulita, M. F. Enrichment of Astrocyte-Derived Extracellular Vesicles from Human Plasma. J. Vis. Exp. (186), e64107, doi:10.3791/64107 (2022).

View Video