Skip to main content
Log in

Platelet-derived extracellular vesicles in Huntington’s disease

  • Original Communication
  • Published:
Journal of Neurology Aims and scope Submit manuscript

Abstract

The production and release of extracellular vesicles (EV) is a property shared by all eukaryotic cells and a phenomenon frequently exacerbated in pathological conditions. The protein cargo of EV, their cell type signature and availability in bodily fluids make them particularly appealing as biomarkers. We recently demonstrated that platelets, among all types of blood cells, contain the highest concentrations of the mutant huntingtin protein (mHtt)—the genetic product of Huntington’s disease (HD), a neurodegenerative disorder which manifests in adulthood with a complex combination of motor, cognitive and psychiatric deficits. Herein, we used a cohort of 59 HD patients at all stages of the disease, including individuals in pre-manifest stages, and 54 healthy age- and sex-matched controls, to evaluate the potential of EV derived from platelets as a biomarker. We found that platelets of pre-manifest and manifest HD patients do not release more EV even if they are activated. Importantly, mHtt was not found within EV derived from platelets, despite them containing high levels of this protein. Correlation analyses also failed to reveal an association between the number of platelet-derived EV and the age of the patients, the number of CAG repeats, the Unified Huntington Disease Rating Scale total motor score, the Total Functional Capacity score or the Burden of Disease score. Our data would, therefore, suggest that EV derived from platelets with HD is not a valuable biomarker in HD.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  1. Manyam BV, Katz L, Hare TA et al (1981) Isoniazid-induced elevation of CSF GABA levels and effects on chorea in Huntington’s disease. Ann Neurol 10:35–37. https://doi.org/10.1002/ana.410100107

    Article  CAS  PubMed  Google Scholar 

  2. Manyam NV, Hare TA, Katz L (1980) Effect of isoniazid on cerebrospinal fluid and plasma GABA levels in Huntington’s disease. Life Sci 26:1303–1308. https://doi.org/10.1016/0024-3205(80)90089-2

    Article  CAS  PubMed  Google Scholar 

  3. Silajdzic E, Rezeli M, Vegvari A et al (2013) A critical evaluation of inflammatory markers in Huntington’s disease plasma. J Huntingt Dis 2:125–134. https://doi.org/10.3233/jhd-130049

    Article  CAS  Google Scholar 

  4. Björkqvist M, Wild EJ, Thiele J et al (2008) A novel pathogenic pathway of immune activation detectable before clinical onset in Huntington’s disease. J Exp Med 205:1869–1877. https://doi.org/10.1084/jem.20080178

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Aronson NN, Blanchard CJ, Madura JD (1997) Homology modeling of glycosyl hydrolase family 18 enzymes and proteins. J Chem Inf Comput Sci 37:999–1005. https://doi.org/10.1021/ci970236v

    Article  CAS  PubMed  Google Scholar 

  6. Vinther-Jensen T, Budtz-Jorgensen E, Simonsen AH et al (2014) YKL-40 in cerebrospinal fluid in Huntington’s disease—a role in pathology or a nonspecific response to inflammation? Parkinsonism Relat Disord 20:1301–1303. https://doi.org/10.1016/j.parkreldis.2014.08.011

    Article  PubMed  Google Scholar 

  7. Georgiou-Karistianis N, Scahill R, Tabrizi SJ et al (2013) Structural MRI in Huntington’s disease and recommendations for its potential use in clinical trials. Neurosci Biobehav Rev 37:480–490. https://doi.org/10.1016/j.neubiorev.2013.01.022

    Article  PubMed  Google Scholar 

  8. Sanchez-Castaneda C, Cherubini A, Elifani F et al (2013) Seeking Huntington disease biomarkers by multimodal, cross-sectional basal ganglia imaging. Hum Brain Mapp 34:1625–1635. https://doi.org/10.1002/hbm.22019

    Article  PubMed  Google Scholar 

  9. Bialas WA, Markesbery WR, Butterfield DA (1980) Increased chloride transport in erythrocytes in Huntington’s disease. Biochem Biophys Res Commun 95:1895–1900. https://doi.org/10.1016/S0006-291X(80)80121-5

    Article  CAS  PubMed  Google Scholar 

  10. Chen C-M, Wu Y-R, Cheng M-L et al (2007) Increased oxidative damage and mitochondrial abnormalities in the peripheral blood of Huntington’s disease patients. Biochem Biophys Res Commun 359:335–340. https://doi.org/10.1016/j.bbrc.2007.05.093

    Article  CAS  PubMed  Google Scholar 

  11. Kraus PH, Vigenschow H, Przuntek H (1986) Spin label study of red blood cell membranes in Huntington’s disease. Eur Neurol 25:61–66. https://doi.org/10.1159/000115988

    Article  CAS  PubMed  Google Scholar 

  12. Zakharov SF, Shandala AM, Shcheglova MV et al (1990) Comparative study of human erythrocyte membranes in normal people and in Huntington’s chorea patients. Vopr Med Khim 36:71–73

    CAS  PubMed  Google Scholar 

  13. Andre R, Carty L, Tabrizi SJ (2016) Disruption of immune cell function by mutant huntingtin in Huntington’s disease pathogenesis. Curr Opin Pharmacol 26:33–38. https://doi.org/10.1016/j.coph.2015.09.008

    Article  CAS  PubMed  Google Scholar 

  14. Cheng M-L, Chang K-H, Wu Y-R, Chen C-M (2016) Metabolic disturbances in plasma as biomarkers for Huntington’s disease. J Nutr Biochem 31:38–44. https://doi.org/10.1016/j.jnutbio.2015.12.001

    Article  CAS  PubMed  Google Scholar 

  15. Narayanan KL, Chopra V, Rosas HD et al (2016) Rho kinase pathway alterations in the brain and leukocytes in Huntington’s disease. Mol Neurobiol 53:2132–2140. https://doi.org/10.1007/s12035-015-9147-9

    Article  CAS  PubMed  Google Scholar 

  16. Träger U, Andre R, Lahiri N et al (2014) HTT-lowering reverses Huntington’s disease immune dysfunction caused by NFκB pathway dysregulation. Brain 137:819–833. https://doi.org/10.1093/brain/awt355

    Article  PubMed  PubMed Central  Google Scholar 

  17. Varani K, Abbracchio MP, Cannella M et al (2003) Aberrant A2A receptor function in peripheral blood cells in Huntington’s disease. FASEB J 17:2148–2150. https://doi.org/10.1096/fj.03-0079fje

    Article  CAS  PubMed  Google Scholar 

  18. Weiss A, Trager U, Wild EJ et al (2012) Mutant huntingtin fragmentation in immune cells tracks Huntington’s disease progression. J Clin Investig 122:3731–3736. https://doi.org/10.1172/jci64565

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Aminoff MJ, Trenchard A, Turner P et al (1974) Plasma uptake of dopamine and 5-hydroxytryptamine and plasma-catecholamine levels in patients with Huntington’s chorea. Lancet 2:1115–1116. https://doi.org/10.1016/S0140-6736(74)90873-3

    Article  CAS  PubMed  Google Scholar 

  20. Diez-Ewald M, Bonilla E, Gonzalez JV (1980) Platelet aggregation, 5-hydroxytryptamine uptake and release in Huntington’s chorea. Prog Neuropsychopharmacol Biol Psychiatry 4:277–283

    Article  CAS  Google Scholar 

  21. Gu M, Gash MT, Mann VM et al (1996) Mitochondrial defect in Huntington’s disease caudate nucleus. Ann Neurol 39:385–389. https://doi.org/10.1002/ana.410390317

    Article  CAS  PubMed  Google Scholar 

  22. Maglione V, Cannella M, Martino T et al (2006) The platelet maximum number of A2A-receptor binding sites (Bmax) linearly correlates with age at onset and CAG repeat expansion in Huntington’s disease patients with predominant chorea. Neurosci Lett 393:27–30. https://doi.org/10.1016/j.neulet.2005.09.037

    Article  CAS  PubMed  Google Scholar 

  23. Markianos M, Panas M, Kalfakis N, Vassilopoulos D (2004) Platelet monoamine oxidase activity in subjects tested for Huntington’s disease gene mutation. J Neural Transm 111:475–483. https://doi.org/10.1007/s00702-003-0103-x

    Article  CAS  PubMed  Google Scholar 

  24. Muramatsu Y, Kaiya H, Imai H et al (1982) Abnormal platelet aggregation response in Huntington’s disease. Arch Psychiatry Neurol Sci (1970) 232:191–200. https://doi.org/10.1007/BF02141780

    Article  CAS  Google Scholar 

  25. Reilmann R, Rolf LH, Lange HW (1994) Huntington’s disease: the neuroexcitotoxin aspartate is increased in platelets and decreased in plasma. J Neurol Sci 127:48–53. https://doi.org/10.1016/0022-510X(94)90134-1

    Article  CAS  PubMed  Google Scholar 

  26. Silva AC, Almeida S, Laço M et al (2013) Mitochondrial respiratory chain complex activity and bioenergetic alterations in human platelets derived from pre-symptomatic and symptomatic Huntington’s disease carriers. Mitochondrion 13:801–809. https://doi.org/10.1016/j.mito.2013.05.006

    Article  CAS  PubMed  Google Scholar 

  27. Tukiainen E, Wikström J, Kilpeläinen H (1981) Uptake of 5-hydroxytryptamine by blood platelets in Huntington’s chorea and Alzheimer type of presenile dementia. Med Biol 59:116–120

    CAS  PubMed  Google Scholar 

  28. Leoni V, Caccia C (2015) The impairment of cholesterol metabolism in Huntington disease. Biochem Biophys Acta 1851:1095–1105. https://doi.org/10.1016/j.bbalip.2014.12.018

    Article  CAS  PubMed  Google Scholar 

  29. Weiss A, Abramowski D, Bibel M et al (2009) Single-step detection of mutant huntingtin in animal and human tissues: a bioassay for Huntington’s disease. Anal Biochem 395:8–15. https://doi.org/10.1016/j.ab.2009.08.001

    Article  CAS  PubMed  Google Scholar 

  30. Anderson AN, Roncaroli F, Hodges A et al (2008) Chromosomal profiles of gene expression in Huntington’s disease. Brain 131:381–388. https://doi.org/10.1093/brain/awm312

    Article  PubMed  Google Scholar 

  31. Borovecki F, Lovrecic L, Zhou J et al (2005) Genome-wide expression profiling of human blood reveals biomarkers for Huntington’s disease. Proc Natl Acad Sci 102:11023–11028. https://doi.org/10.1073/pnas.0504921102

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Aylward EH, Nopoulos PC, Ross CA et al (2011) Longitudinal change in regional brain volumes in prodromal Huntington disease. J Neurol Neurosurg Psychiatry 82:405–410. https://doi.org/10.1136/jnnp.2010.208264

    Article  PubMed  Google Scholar 

  33. Bohanna I, Georgiou-Karistianis N, Sritharan A et al (2011) Diffusion tensor imaging in Huntington’s disease reveals distinct patterns of white matter degeneration associated with motor and cognitive deficits. Brain Imaging Behav 5:171–180. https://doi.org/10.1007/s11682-011-9121-8

    Article  PubMed  Google Scholar 

  34. Davie CA, Barker GJ, Quinn N et al (1994) Proton MRS in Huntington’s disease. Lancet 343:1580. https://doi.org/10.1016/S0140-6736(94)92987-4

    Article  CAS  PubMed  Google Scholar 

  35. Jenkins BG, Rosas HD, Chen YC et al (1998) 1H NMR spectroscopy studies of Huntington’s disease: correlations with CAG repeat numbers. Neurology 50:1357–1365

    Article  CAS  PubMed  Google Scholar 

  36. Paulsen JS, Zimbelman JL, Hinton SC et al (2004) fMRI biomarker of early neuronal dysfunction in presymptomatic Huntington’s disease. Am J Neuroradiol 25:1715–1721

    PubMed  PubMed Central  Google Scholar 

  37. Rosas HD, Chen YI, Doros G et al (2012) Alterations in brain transition metals in Huntington disease: an evolving and intricate story. Arch Neurol 69:887–893. https://doi.org/10.1001/archneurol.2011.2945

    Article  PubMed  PubMed Central  Google Scholar 

  38. Ruocco HH, Bonilha L, Li LM et al (2008) Longitudinal analysis of regional grey matter loss in Huntington disease: effects of the length of the expanded CAG repeat. J Neurol Neurosurg Psychiatry 79:130–135. https://doi.org/10.1136/jnnp.2007.116244

    Article  CAS  PubMed  Google Scholar 

  39. Shin H, Kim MH, Lee SJ et al (2013) Decreased metabolism in the cerebral cortex in early-stage Huntington’s disease: a possible biomarker of disease progression? J Clin Neurosci 9:21–25. https://doi.org/10.3988/jcn.2013.9.1.21

    Article  Google Scholar 

  40. Sritharan A, Egan GF, Johnston L et al (2010) A longitudinal diffusion tensor imaging study in symptomatic Huntington’s disease. J Neurol Neurosurg Psychiatry 81:257–262. https://doi.org/10.1136/jnnp.2007.142786

    Article  PubMed  Google Scholar 

  41. Sturrock A, Laule C, Decolongon J et al (2010) Magnetic resonance spectroscopy biomarkers in premanifest and early Huntington disease. Neurology 75:1702–1710. https://doi.org/10.1212/WNL.0b013e3181fc27e4

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Disatnik M-H, Joshi AU, Saw NL et al (2016) Potential biomarkers to follow the progression and treatment response of Huntington’s disease. J Exp Med 213:2655–2669. https://doi.org/10.1084/jem.20160776

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Olsson MG, Davidsson S, Muhammad ZD et al (2012) Increased levels of hemoglobin and alpha1-microglobulin in Huntington’s disease. Front Biosci (Elite Ed) 4:950–957

    Google Scholar 

  44. Wood NI, Goodman AOG, van der Burg JMM et al (2008) Increased thirst and drinking in Huntington’s disease and the R6/2 mouse. Brain Res Bull 76:70–79. https://doi.org/10.1016/j.brainresbull.2007.12.007

    Article  CAS  PubMed  Google Scholar 

  45. Byrne LM, Wild EJ (2016) Cerebrospinal fluid biomarkers for Huntington’s disease. J Huntingt Dis 5:1–13. https://doi.org/10.3233/jhd-160196

    Article  CAS  Google Scholar 

  46. Chen X, Guo C, Kong J (2012) Oxidative stress in neurodegenerative diseases. Neural Regener Res 7:376–385. https://doi.org/10.3969/j.issn.1673-5374.2012.05.009

    Article  CAS  Google Scholar 

  47. Garrett MC, Soares-da-Silva P (1992) Increased cerebrospinal fluid dopamine and 3,4-dihydroxyphenylacetic acid levels in Huntington’s disease: evidence for an overactive dopaminergic brain transmission. J Neurochem 58:101–106. https://doi.org/10.1111/j.1471-4159.1992.tb09283.x

    Article  CAS  PubMed  Google Scholar 

  48. Kurlan R, Caine E, Rubin A et al (1988) Cerebrospinal fluid correlates of depression in Huntington’s disease. Arch Neurol 45:881–883. https://doi.org/10.1001/archneur.1988.00520320071018

    Article  CAS  PubMed  Google Scholar 

  49. Manyam BV, Giacobini E, Colliver JA (1990) Cerebrospinal fluid acetylcholinesterase and choline measurements in Huntington’s disease. J Neurol 237:281–284. https://doi.org/10.1007/BF00314742

    Article  CAS  PubMed  Google Scholar 

  50. Vinther-Jensen T, Simonsen AH, Budtz-Jørgensen E et al (2015) Ubiquitin: a potential cerebrospinal fluid progression marker in Huntington’s disease. Eur J Neurol 22:1378–1384. https://doi.org/10.1111/ene.12750

    Article  CAS  PubMed  Google Scholar 

  51. Carrizzo A, Di Pardo A, Maglione V et al (2014) Nitric oxide dysregulation in platelets from patients with advanced Huntington disease. PLoS One 9:e89745. https://doi.org/10.1371/journal.pone.0089745

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Byrne LM, Rodrigues FB, Blennow K et al (2017) Neurofilament light protein in blood as a potential biomarker of neurodegeneration in Huntington’s disease: a retrospective cohort analysis. Lancet Neurol. https://doi.org/10.1016/s1474-4422(17)30124-2

    Article  PubMed  PubMed Central  Google Scholar 

  53. Vinther-Jensen T, Bornsen L, Budtz-Jorgensen E et al (2016) Selected CSF biomarkers indicate no evidence of early neuroinflammation in Huntington disease. Neurol Neuroimmunol Neuroinflamm 3:e287. https://doi.org/10.1212/nxi.0000000000000287

    Article  PubMed  PubMed Central  Google Scholar 

  54. Tan Z, Dai W, van Erp TG et al (2015) Huntington’s disease cerebrospinal fluid seeds aggregation of mutant huntingtin. Mol Psychiatry 20:1286–1293. https://doi.org/10.1038/mp.2015.81

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Wild EJ, Boggio R, Langbehn D et al (2015) Quantification of mutant huntingtin protein in cerebrospinal fluid from Huntington’s disease patients. J Clin Investig 125:1979–1986. https://doi.org/10.1172/JCI80743

    Article  PubMed  PubMed Central  Google Scholar 

  56. Marcoux G, Duchez A-C, Cloutier N et al (2016) Revealing the diversity of extracellular vesicles using high-dimensional flow cytometry analyses. Sci Rep 6:35928. https://doi.org/10.1038/srep35928

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. van der Pol E, Boing AN, Gool EL, Nieuwland R (2016) Recent developments in the nomenclature, presence, isolation, detection and clinical impact of extracellular vesicles. J Thromb Haemost JTH 14:48–56. https://doi.org/10.1111/jth.13190

    Article  PubMed  Google Scholar 

  58. György B, Szabó TG, Pásztói M et al (2011) Membrane vesicles, current state-of-the-art: emerging role of extracellular vesicles. Cell Mol Life Sci 68:2667–2688. https://doi.org/10.1007/s00018-011-0689-3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. EL Andaloussi S, Mäger I, Breakefield XO, Wood MJA (2013) Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov 12:347–357. https://doi.org/10.1038/nrd3978

    Article  CAS  Google Scholar 

  60. Lötvall J, Hill AF, Hochberg F et al (2014) Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles. J Extracell Vesicles 3:26913. https://doi.org/10.3402/jev.v3.26913

    Article  PubMed  Google Scholar 

  61. Raposo G, Stoorvogel W (2013) Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol 200:373–383. https://doi.org/10.1083/jcb.201211138

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Cloutier N, Tan S, Boudreau LH et al (2013) The exposure of autoantigens by microparticles underlies the formation of potent inflammatory components: the microparticle-associated immune complexes. EMBO Mol Med 5:235–249. https://doi.org/10.1002/emmm.201201846

    Article  CAS  PubMed  Google Scholar 

  63. Inal JM, Fairbrother U, Heugh S (2013) Microvesiculation and disease. Biochem Soc Trans 41:237–240. https://doi.org/10.1042/bst20120258

    Article  CAS  PubMed  Google Scholar 

  64. Denis HL, Lamontagne-Proulx J, St-Amour I et al (2018) Platelet abnormalities in Huntington’s disease. J Neurol Neurosurg Psychiatry (in revision)

  65. Rousseau M, Belleannee C, Duchez A-C et al (2015) Detection and quantification of microparticles from different cellular lineages using flow cytometry. Evaluation of the impact of secreted phospholipase A2 on microparticle assessment. PLoS One 10:e0116812. https://doi.org/10.1371/journal.pone.0116812

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Duchez A-C, Boudreau LH, Naika GS et al (2015) Platelet microparticles are internalized in neutrophils via the concerted activity of 12-lipoxygenase and secreted phospholipase A2-IIA. Proc Natl Acad Sci 112:E3564–E3573. https://doi.org/10.1073/pnas.1507905112

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Boudreau LH, Duchez A-C, Cloutier N et al (2014) Platelets release mitochondria serving as substrate for bactericidal group IIA-secreted phospholipase A2 to promote inflammation. Blood 124:2173–2183. https://doi.org/10.1182/blood-2014-05-573543

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Lamontagne-Proulx J, St-Amour I, Labib R et al (2018) Erythrocyte-derived extracellular vesicles: a novel, robust and specific biomarker that maps to Parkinson’s disease stages. Neuro Dis (Under review)

Download references

Acknowledgements

The study was funded by an operating grant from the Merck Sharpe & Dohme to F. C. who is also a recipient of a Research Chair from the Fonds de Recherche du Québec en santé (FRQS) providing salary support and operating funds. I. S.-A. was supported by a CIHR-Huntington Society of Canada postdoctoral fellowship. R. A. B. and S. L. M. are supported by a National Institute for Health Research (NIHR) award of a Biomedical Research Center to the University of Cambridge and Addenbrooke’s Hospital. E. B. is supported by the Canadian Institutes of Health Research. N. D. MD-MSc. also funded by CIHR and by Canadian Consortium on Neurodegeneration in Aging (CCNA). HLD and JPL hold a Desjardins scholarship from the Fondation du CHU de Québec. HLD hold a bourse d’excellence du Centre Thématique de Recherche en Neurosciences (CTRN) du CHU de Québec. The authors would like to thank all the students and staff who helped with the blood collections in Cambridge, Quebec City and Montreal and importantly, all patients and their families for being so generous with their time for participating in this study.

Author information

Authors and Affiliations

Authors

Contributions

HLD participated in experiments, data analysis/interpretation and preparation of figures. She wrote the first draft of the manuscript and helped with subsequent revisions. JLP participated in the design of the experiments and various aspects of the study including blood collections, experiments and data analysis. IS-A participated in the design of the experiments and blood collections, took part in some data analysis and interpretation. SLM helped with patient recruitment in Cambridge and participated in the preparation of blood collection. AW performed T-FRET analyses related to Fig. 1i. SC recruited patients in Montreal. RAB recruited patients in Cambridge, participated in data interpretation and revised the manuscript. EB initiated the study and was involved in the experimental design. He also revised the manuscript. FC initiated the study and was involved in the experimental design. She supervised the project and wrote the manuscript.

Corresponding authors

Correspondence to Eric Boilard or Francesca Cicchetti.

Ethics declarations

Conflicts of interest

The authors declare that they have no competing interests.

Electronic supplementary material

Below is the link to the electronic supplementary material.

415_2018_9022_MOESM1_ESM.pdf

Supplementary material 1 Figure S1. Identified biomarkers in HD. Summary of the literature for all reported biomarkers of HD in CSF, blood, urine and brain. Abbreviations: DOPA: 3,4-dihydroxyphenylalanine; DOPAC: 3,4-dihydroxyphenylacetic acid; IL-6: Interleukin 6;IL-8: Interleukin 8; NMDA: N-methyl-D-aspartate; mHtt: mutant huntingtin; Cu/Zn-SOD: Cu/Zn Superoxide Dismutase; HD: Huntington disease; PCYT1A: Phosphate Cytidylyltransferase 1, Pre-HD : pre-manifest, Choline, Alpha; YKL-40: chitinase-like protein-40; 5-HIAA: 5-hydroxyindoleacetic acid; 8-OHdG: 8-hydroxy-2’ -deoxyguanosine; 8-oxodG6: 8-oxo-7,8-dihydro-2′-deoxyguanosine; ↑ ; increase; ↓; decrease; ✔; presence (PDF 13620 KB)

415_2018_9022_MOESM2_ESM.pdf

Supplementary material 2 Figure S2. (A) Absence of statistical differences in comorbidities of healthy control patients - which including depression (p=0.2455), diabetes (p=0.0749), hypertension (p=0.9664), hypercholesterolemia (p=0.3615), allergies (p=0.9904) and anxiety (p=0.4675) - and counts of EV derived from platelets. EV from patients without comorbidities were further compared to patients with one or more comorbidities. Again, no statistically significant differences were found (p=0.6745). Statistical analyses were performed using the non-parametric Mann Whitney test. (B) Immunoblot of CD41a, ALIX, TSG101, actin and VDAC in platelet-derived EV or plts. Data are representative of 3 independent experiments. (C) Left panel: After the acquisition of fluorescent signals, an initial gating was performed on all data to exclude counting beads from files. In this experiment, 144 beads were counted. Right panel: Representation of SSC-H (granularity) and FSC-PMT-H (relative size) dot plots of platelet-derived EV in PFP detecting using PerCP-CyTM5.5-conjugated annexin V and V450-conjugated antibodies directed against CD41. The size of platelet-derived EV ranged between 100 and 1000nm. Abbreviations: ALIX, programmed cell death 6 interacting protein; EV, extracellular vesicles; plts, platelets; TSG101, tumor susceptibility gene 101 protein; VDAC, Voltage-dependent anion-selective channel 1 (PDF 1813 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Denis, H.L., Lamontagne-Proulx, J., St-Amour, I. et al. Platelet-derived extracellular vesicles in Huntington’s disease. J Neurol 265, 2704–2712 (2018). https://doi.org/10.1007/s00415-018-9022-5

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00415-018-9022-5

Keywords

Navigation