2,546
Views
20
CrossRef citations to date
0
Altmetric
Reviews

Blood cell-derived extracellular vesicles: diagnostic biomarkers and smart delivery systems

, , , , , & show all
Pages 7929-7940 | Received 14 Jul 2021, Accepted 12 Sep 2021, Published online: 08 Oct 2021

References

  • van Niel G, D’Angelo G, Raposo G, Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19(4):213–228.
  • Zaborowski MP, Balaj L, Breakefield XO, Lai CP Extracellular vesicles: composition, biological relevance, and methods of study. BIOSCIENCE. 2015;65(8):783–797.
  • Yanez-Mo M, Siljander PR, Andreu Z, et al. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles. 2015;4:27066.
  • Jayaseelan VP. Emerging role of exosomes as promising diagnostic tool for cancer. CANCER GENE THER. 2020;27(6):395–398.
  • Moller A. Lobb RJ The evolving translational potential of small extracellular vesicles in cancer. NAT REV CANCER. 2020. DOI:10.1038/s41568-020-00299-w
  • O’Brien K, Breyne K, Ughetto S, Breakefield XO et al. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat Rev Mol Cell Biol. 2020;21(10):585–606.
  • de Jong OG, Kooijmans S, Murphy DE, Schiffelers RM, et al. Drug delivery with extracellular vesicles: from imagination to innovation. Acc Chem Res. 2019;52(7):1761–1770.
  • AS Mager, I, Breakefield XO, Wood MJ: Extracellular vesicles: biology and emerging therapeutic opportunities. Nature Reviews Drug Discovery. 2013;12(5):347–357.
  • Shi J, Kundrat L, Pishesha N, Lodish HF, et al. Engineered red blood cells as carriers for systemic delivery of a wide array of functional probes. Proc Natl Acad Sci U S A. 2014;111(28):10131–10136.
  • Rubin O, Delobel J, Prudent M, Angelillo-Scherrer A, et al. Red blood cell-derived microparticles isolated from blood units initiate and propagate thrombin generation. TRANSFUSION. 2013;53(8):1744–1754.
  • Crawford TM, Andersen CC, Hodyl NA, Stark MJ, et al. The contribution of red blood cell transfusion to neonatal morbidity and mortality. J Paediatr Child Health. 2019;55(4):387–392.
  • Connor JP, O’Shea A, McCool K, Barroilhet LM, et al. Peri-operative allogeneic blood transfusion is associated with poor overall survival in advanced epithelial ovarian Cancer; potential impact of patient blood management on Cancer outcomes. GYNECOL ONCOL. 2018;151(2):294–298.
  • Park SY, Seo KS. Karm MH Perioperative red blood cell transfusion in orofacial surgery. J Dent Anesth Pain Med. 2017;17(3):163–181.
  • Danesh A, Inglis HC, Jackman RP, Norris PJ, et al. Exosomes from red blood cell units bind to monocytes and induce proinflammatory cytokines, boosting T-cell responses in vitro. BLOOD. 2014;123(5):687–696.
  • Gao Y, Jin H, Tan H, et al. The role of extracellular vesicles from stored RBC units in B lymphocyte survival and plasma cell differentiation. J Leukoc Biol. 2020. DOI:10.1002/JLB.1A0220-666R
  • Straat M, Boing AN, Tuip-De BA, et al. Extracellular Vesicles from Red Blood Cell Products Induce a Strong Pro-Inflammatory Host Response, Dependent on Both Numbers and Storage Duration. Transfus Med Hemother. 2016;43(4):302–305.
  • van Manen L, Peters AL, van der Sluijs PM, et al. Clearance and phenotype of extracellular vesicles after red blood cell transfusion in a human endotoxemia model. TRANSFUS APHER SCI. 2019;58(4):508–511.
  • Huang H, Zhu J, Fan L, et al. MicroRNA Profiling of Exosomes Derived from Red Blood Cell Units: implications in Transfusion-Related Immunomodulation. BIOMED RES INT. 2019;2019:2045915.
  • Sheng L, Stewart T, Yang D, et al. Erythrocytic alpha-synuclein contained in microvesicles regulates astrocytic glutamate homeostasis: a new perspective on Parkinson’s disease pathogenesis. Acta Neuropathol Commun. 2020;8(1):102.
  • Klyachko NL, Arzt CJ, Li SM, et al. Extracellular Vesicle-Based Therapeutics: preclinical and Clinical Investigations. PHARMACEUTICS. 2020;12:12.
  • Wang D, Tai P, Gao G. Adeno-associated virus vector as a platform for gene therapy delivery. NAT REV DRUG DISCOV. 2019;18(5):358–378.
  • Kauffman KJ, Dorkin JR, Yang JH, Anderson DG, et al. Optimization of lipid nanoparticle formulations for mrna delivery in vivo with fractional factorial and definitive screening designs. NANO LETT. 2015;15(11):7300–7306.
  • Kanasty R, Dorkin JR, Vegas A, et al. Delivery materials for siRNA therapeutics. NAT MATER. 2013;12(11):967–977.
  • Saint-Pol J, Gosselet F, Duban-Deweer S, et al. Targeting and Crossing the Blood-Brain Barrier with Extracellular Vesicles. CELLS-BASEL. 2020;9:4.
  • Kibria G, Ramos EK, Wan Y, et al. Exosomes as a Drug Delivery System in Cancer Therapy: potential and Challenges. Mol Pharm. 2018;15(9):3625–3633.
  • Barile L, Vassalli G. Exosomes: therapy delivery tools and biomarkers of diseases. Pharmacol Ther. 2017;174:63–78.
  • Duan L, Xu L, Xu X, et al. Exosome-mediated delivery of gene vectors for gene therapy. NANOSCALE. 2021;13(3):1387–1397.
  • Usman WM, Pham TC, Kwok YY, et al. Efficient RNA drug delivery using red blood cell extracellular vesicles. NAT COMMUN. 2018;9(1):2359.
  • Borgheti-Cardoso LN, Kooijmans S, Chamorro LG, et al. Extracellular vesicles derived from Plasmodium-infected and non-infected red blood cells as targeted drug delivery vehicles. Int J Pharm. 2020;587:119627.
  • Sender R, Fuchs S, Milo R. Revised estimates for the number of human and bacteria cells in the body. PLOS BIOL. 2016;14(8):e1002533.
  • Bekeschus S, Seebauer C, Wende K, et al. Physical plasma and leukocytes - immune or reactive? BIOL CHEM. 2018;400(1):63–75.
  • Pugholm LH, Baek R, Sondergaard EK, et al. Phenotyping of Leukocytes and Leukocyte-Derived Extracellular Vesicles. J IMMUNOL RES. 2016;2016:6391264.
  • Gasser O, Hess C, Miot S, et al. Characterisation and properties of ectosomes released by human polymorphonuclear neutrophils. EXP CELL RES. 2003;285(2):243–257.
  • Danesh A, Inglis HC, Abdel-Mohsen M, et al. Granulocyte-Derived Extracellular Vesicles Activate Monocytes and Are Associated With Mortality in Intensive Care Unit Patients. FRONT IMMUNOL. 2018;9:956.
  • Gasser O, Schifferli JA. Activated polymorphonuclear neutrophils disseminate anti-inflammatory microparticles by ectocytosis. BLOOD. 2004;104(8):2543–2548.
  • Xie Y, Zhang X, Zhao T, et al. Natural CD8(+)25(+) regulatory T cell-secreted exosomes capable of suppressing cytotoxic T lymphocyte-mediated immunity against B16 melanoma. Biochem Biophys Res Commun. 2013;438(1):152–155.
  • Tung SL, Boardman DA, Sen M, et al. Regulatory T cell-derived extracellular vesicles modify dendritic cell function. Sci Rep. 2018;8(1):6065.
  • Zhang F, Li R, Yang Y, et al. Specific Decrease in B-Cell-Derived Extracellular Vesicles Enhances Post-Chemotherapeutic CD8(+) T Cell Responses. IMMUNITY. 2019;50(3):738–750.
  • Meyer AD, Rishmawi AR, Kamucheka R, et al. Effect of blood flow on platelets, leukocytes, and extracellular vesicles in thrombosis of simulated neonatal extracorporeal circulation. J THROMB HAEMOST. 2020;18(2):399–410.
  • Chennakrishnaiah S, Meehan B, D’Asti E, et al. Leukocytes as a reservoir of circulating oncogenic DNA and regulatory targets of tumor-derived extracellular vesicles. J THROMB HAEMOST. 2018;16(9):1800–1813.
  • Banks WA, Gray AM, Erickson MA, et al. Lipopolysaccharide-induced blood-brain barrier disruption: roles of cyclooxygenase, oxidative stress, neuroinflammation, and elements of the neurovascular unit. J Neuroinflammation. 2015;12:223.
  • Sarlon-Bartoli G, Bennis Y, Lacroix R, et al. Plasmatic level of leukocyte-derived microparticles is associated with unstable plaque in asymptomatic patients with high-grade carotid stenosis. J AM COLL CARDIOL. 2013;62(16):1436–1441.
  • Nguyen MA, Karunakaran D, Geoffrion M, et al. Extracellular Vesicles Secreted by Atherogenic Macrophages Transfer MicroRNA to Inhibit Cell Migration. Arterioscler Thromb Vasc Biol. 2018;38(1):49–63.
  • Cognasse F, Laradi S, Berthelot P, et al. Platelet Inflammatory Response to Stress. FRONT IMMUNOL. 2019;10:1478.
  • van der Meijden P, Heemskerk J. Platelet biology and functions: new concepts and clinical perspectives. NAT REV CARDIOL. 2019;16(3):166–179.
  • Mitrugno A, Tassi YS, Sylman JL, et al. The role of coagulation and platelets in colon cancer-associated thrombosis. Am J Physiol Cell Physiol. 2019;316(2):C264–C273.
  • Zubairova LD, Nabiullina RM, Nagaswami C, et al. Circulating Microparticles Alter Formation, Structure, and Properties of Fibrin Clots. Sci Rep. 2015;5:17611.
  • Dyer MR, Alexander W, Hassoune A, et al. Platelet-derived extracellular vesicles released after trauma promote hemostasis and contribute to DVT in mice. J THROMB HAEMOST. 2019;17(10):1733–1745.
  • Marcoux G, Magron A, Sut C, et al. Platelet-derived extracellular vesicles convey mitochondrial DAMPs in platelet concentrates and their levels are associated with adverse reactions. TRANSFUSION. 2019;59(7):2403–2414.
  • Kapur R, Zufferey A, Boilard E, et al. Nouvelle cuisine: platelets served with inflammation. J IMMUNOL. 2015;194(12):5579–5587.
  • Chimen M, Evryviadou A, Box CL, et al. Appropriation of GPIbalpha from platelet-derived extracellular vesicles supports monocyte recruitment in systemic inflammation. HAEMATOLOGICA. 2020;105(5):1248–1261.
  • Dinkla S, van Cranenbroek B, van der Heijden WA, et al. Platelet microparticles inhibit IL-17 production by regulatory T cells through P-selectin. BLOOD. 2016;127(16):1976–1986.
  • Sprague DL, Elzey BD, Crist SA, et al. Platelet-mediated modulation of adaptive immunity: unique delivery of CD154 signal by platelet-derived membrane vesicles. BLOOD. 2008;111(10):5028–5036.
  • Chiva-Blanch G, Padro T, Alonso R, et al. Liquid Biopsy of Extracellular Microvesicles Maps Coronary Calcification and Atherosclerotic Plaque in Asymptomatic Patients With Familial Hypercholesterolemia. Arterioscler Thromb Vasc Biol. 2019;39(5):945–955.
  • Fortin PR, Cloutier N, Bissonnette V, et al. Distinct Subtypes of microparticle-containing immune complexes are associated with disease activity, damage, and carotid intima-media thickness in systemic lupus erythematosus. J RHEUMATOL. 2016;43(11):2019–2025.
  • Helmke A, von Vietinghoff S. Extracellular vesicles as mediators of vascular inflammation in kidney disease. World J Nephrol. 2016;5(2):125–138.
  • Tessandier N, Melki I, Cloutier N, et al. Platelets disseminate extracellular vesicles in lymph in rheumatoid arthritis. Arterioscler Thromb Vasc Biol. 2020;40(4):929–942.
  • Schrottmaier WC, Mussbacher M, Salzmann M, et al. Platelet-leukocyte interplay during vascular disease. ATHEROSCLEROSIS. 2020;307:109–120.
  • Ma Q, Fan Q, Xu J, et al. Calming cytokine storm in pneumonia by targeted delivery of TPCA-1 using platelet-derived extracellular vesicles. Matter. 2020;3(1):287–301.
  • Dovizio M, Bruno A, Contursi A, et al. Platelets and extracellular vesicles in cancer: diagnostic and therapeutic implications. Cancer Metastasis Rev. 2018;37(2–3):455–467.
  • Zmigrodzka M, Witkowska-Pilaszewicz O, Winnicka A. Platelets Extracellular Vesicles as Regulators of Cancer Progression-An Updated Perspective. INT J MOL SCI. 2020;21:15.
  • Kikuchi S, Yoshioka Y, Prieto-Vila M, et al. Involvement of Extracellular Vesicles in Vascular-Related Functions in Cancer Progression and Metastasis. INT J MOL SCI. 2019;20:10.
  • Sun Y, Liu XL, Zhang D, et al. Platelet-derived exosomes affect the proliferation and migration of human umbilical vein endothelial cells Via miR-126. CURR VASC PHARMACOL. 2019;17(4):379–387.
  • Gaetani E, Del ZF, Marcantoni M, et al. Microparticles produced by activated platelets carry a potent and functionally active angiogenic signal in subjects with crohn’s disease. INT J MOL SCI. 2018;19:10.
  • Alberro A, Osorio-Querejeta I, Sepulveda L, et al. T cells and immune functions of plasma extracellular vesicles are differentially modulated from adults to centenarians. Aging (Albany NY). 2019;11(22):10723–10741.
  • Bank IE, Timmers L, Gijsberts CM, et al. The diagnostic and prognostic potential of plasma extracellular vesicles for cardiovascular disease. EXPERT REV MOL DIAGN. 2015;15(12):1577–1588.
  • Boyiadzis M, Whiteside TL. Plasma-derived exosomes in acute myeloid leukemia for detection of minimal residual disease: are we ready? EXPERT REV MOL DIAGN. 2016;16(6):623–629.
  • Cha DJ, Mengel D, Mustapic M, et al. miR-212 and miR-132 are downregulated in neurally derived plasma exosomes of alzheimer’s patients. Front Neurosci. 2019;13:1208.
  • Ohmichi T, Mitsuhashi M, Tatebe H, et al. Quantification of brain-derived extracellular vesicles in plasma as a biomarker to diagnose Parkinson’s and related diseases. Parkinsonism Relat Disord. 2019;61:82–87.
  • Shi M, Liu C, Cook TJ, et al. Plasma exosomal alpha-synuclein is likely CNS-derived and increased in Parkinson’s disease. ACTA NEUROPATHOL. 2014;128(5):639–650.
  • Wang J, Liu Y, Sun W, et al. Plasma exosomes as novel biomarker for the early diagnosis of gastric cancer. CANCER BIOMARK. 2018;21(4):805–812.
  • Hannafon BN, Trigoso YD, Calloway CL, et al. Plasma exosome microRNAs are indicative of breast cancer. BREAST CANCER RES. 2016;18(1):90.
  • Joncas FH, Lucien F, Rouleau M, et al. Plasma extracellular vesicles as phenotypic biomarkers in prostate cancer patients. PROSTATE. 2019;79(15):1767–1776.
  • Hoshino A, Kim HS, Bojmar L, et al. Extracellular vesicle and particle biomarkers define multiple human cancers. CELL. 2020;182(4):1044–1061.
  • Pomatto M, Bussolati B, D’Antico S, et al. Improved loading of plasma-derived extracellular vesicles to encapsulate antitumor miRNAs. Mol Ther Methods Clin Dev. 2019;13:133–144.
  • Wahlgren J, L Kt D, Brisslert M, et al. Plasma exosomes can deliver exogenous short interfering RNA to monocytes and lymphocytes. NUCLEIC ACIDS RES. 2012;40(17):e130.
  • Xu X, Liang Y, Li X, et al. Exosome-mediated delivery of kartogenin for chondrogenesis of synovial fluid-derived mesenchymal stem cells and cartilage regeneration. BIOMATERIALS. 2021;269:120539.
  • Liang Y, Xu X, Li X, et al. Chondrocyte-targeted microrna delivery by engineered exosomes toward a cell-free osteoarthritis therapy. ACS Appl Mater Interfaces. 2020;12(33):36938–36947.
  • Liang Y, Duan L, Lu J, et al. Engineering exosomes for targeted drug delivery. THERANOSTICS. 2021;11(7):3183–3195.
  • Smyth T, Petrova K, Payton NM, et al. Surface functionalization of exosomes using click chemistry. Bioconjug Chem. 2014;25(10):1777–1784.
  • Ye Z, Zhang T, He W, et al. Methotrexate-loaded extracellular vesicles functionalized with therapeutic and targeted peptides for the treatment of glioblastoma multiforme. ACS Appl Mater Interfaces. 2018;10(15):12341–12350.
  • Gao X, Ran N, Dong X, et al. Anchor peptide captures, targets, and loads exosomes of diverse origins for diagnostics and therapy. SCI TRANSL MED. 2018;10:444.
  • Pham TC, Jayasinghe MK, Pham TT, et al. Covalent conjugation of extracellular vesicles with peptides and nanobodies for targeted therapeutic delivery. J Extracell Vesicles. 2021;10(4):e12057.
  • Sidhom K, Obi PO, Saleem A. A review of exosomal isolation methods: is size exclusion chromatography the best option? International Journal of Molecular Sciences. 2020;21(18):18.
  • Li P, Kaslan M, Lee SH, et al. Progress in Exosome Isolation Techniques. THERANOSTICS. 2017;7(3):789–804.
  • Greening DW, Xu R, Ji H, et al. A protocol for exosome isolation and characterization: evaluation of ultracentrifugation, density-gradient separation, and immunoaffinity capture methods. Methods Mol Biol. 2015;1295:179–209.
  • Konoshenko MY, Lekchnov EA, Vlassov AV, et al. Isolation of extracellular vesicles: general methodologies and latest trends. BIOMED RES INT. 2018;2018:8545347.
  • Shirejini SZ, Inci F. The Yin and Yang of exosome isolation methods: conventional practice, microfluidics, and commercial kits. Biotechnology Advances. 2021;107814. DOI:10.1016/j.biotechadv.2021.107814
  • Kang M, Jordan V, Blenkiron C, et al. Biodistribution of extracellular vesicles following administration into animals: a systematic review. J Extracell Vesicles. 2021;10(8):e12085.