Extracellular Vesicles in Haematological Disorders: A Friend or a Foe?
Abstract
:1. Introduction
2. Haematological Malignancies
2.1. Myeloid Disorders
2.1.1. Myelodysplastic Syndromes
2.1.2. Acute Myeloid Leukaemia
2.1.3. Myeloproliferative Disorders
2.2. Lymphoid Disorders
2.2.1. Acute Lymphoblastic Leukaemia
2.2.2. Chronic Lymphocytic Leukaemia
2.2.3. Lymphomas
2.2.4. Multiple Myeloma
3. Conclusions
Funding
Institutional Review Board Statement
Informed Consent Statement
Conflicts of Interest
References
- Boukouris, S.; Mathivanan, S. Exosomes in bodily fluids are a highly stable resource of disease biomarkers. Proteom. Clin. Appl. 2015, 9, 358–367. [Google Scholar] [CrossRef] [PubMed]
- Yuana, Y.; Sturk, A.; Nieuwland, R. Extracellular vesicles in physiological and pathological conditions. Blood Rev. 2013, 27, 31–39. [Google Scholar] [CrossRef] [PubMed]
- Xu, R.; Rai, A.; Chen, M.; Suwakulsiri, W.; Greening, D.W.; Simpson, R.J. Extracellular vesicles in cancer—Implications for future improvements in cancer care. Nat. Rev. Clin. Oncol. 2018, 15, 617–638. [Google Scholar] [CrossRef] [PubMed]
- Russell, A.E.; Sneider, A.; Witwer, K.W.; Bergese, P.; Bhattacharyya, S.N.; Cocks, A.; Cocucci, E.; Erdbrügger, U.; Falcon-Perez, J.M.; Freeman, D.W.; et al. Biological membranes in EV biogenesis, stability, uptake, and cargo transfer: An ISEV position paper arising from the ISEV membranes and EVs workshop. J. Extracell. Vesicles 2019, 8, 1684862. [Google Scholar] [CrossRef]
- Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; 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, 7, 1535750. [Google Scholar] [CrossRef]
- McKelvey, K.J.; Powell, K.L.; Ashton, A.W.; Morris, J.M.; McCracken, S.A. Exosomes: Mechanisms of Uptake. J. Circ. Biomark. 2015, 4, 7. [Google Scholar] [CrossRef]
- Hrustincova, A.; Krejcik, Z.; Kundrat, D.; Szikszai, K.; Belickova, M.; Pecherkova, P.; Klema, J.; Vesela, J.; Hruba, M.; Cermak, J.; et al. Circulating Small Noncoding RNAs Have Specific Expression Patterns in Plasma and Extracellular Vesicles in Myelodysplastic Syndromes and Are Predictive of Patient Outcome. Cells 2020, 9, 794. [Google Scholar] [CrossRef]
- Cerisoli, S.; Busilacchi, E.M.; Mattiucci, D.; Rossi, E.; Mariani, M.; Guescini, M.; Pugnaloni, A.; Olivieri, F.; Olivieri, A.; Poloni, A. The exosomal surface phenotype and inflamma-miR cargo correlate with MDS diagnosis. Br. J. Haematol. 2021, 192, e4–e7. [Google Scholar] [CrossRef]
- Waisbren, J.; Dinner, S.; Altman, J.; Frankfurt, O.; Helenowski, I.; Gao, J.; McMahon, B.J.; Stein, B.L. Disease characteristics and prognosis of myelodysplastic syndrome presenting with isolated thrombocytopenia. Int. J. Hematol. 2016, 105, 44–51. [Google Scholar] [CrossRef]
- Fang, Z.; Wang, X.; Wu, J.; Xiao, R.; Liu, J. High serum extracellular vesicle miR-10b expression predicts poor prognosis in patients with acute myeloid leukemia. Cancer Biomark. 2020, 27, 1–9. [Google Scholar] [CrossRef]
- Hornick, N.I.; Doron, B.; Abdelhamed, S.; Huan, J.; Harrington, C.A.; Shen, R.; Cambronne, X.A.; Verghese, S.C.; Kurre, P. AML suppresses hematopoiesis by releasing exosomes that contain microRNAs targeting c-MYB. Sci. Signal. 2016, 9, ra88. [Google Scholar] [CrossRef] [PubMed]
- Ji, D.; He, Y.; Lu, W.; Rong, Y.; Li, F.; Huang, X.; Huang, R.; Jiang, Y.; Chen, G. Small-sized extracellular vesicles (EVs) derived from acute myeloid leukemia bone marrow mesenchymal stem cells transfer miR-26a-5p to promote acute myeloid leukemia cell proliferation, migration, and invasion. Hum. Cell 2021, 34, 965–976. [Google Scholar] [CrossRef] [PubMed]
- Wang, D.; Ming, X.; Xu, J.; Xiao, Y. Circ_0009910 shuttled by exosomes regulates proliferation, cell cycle and apoptosis of acute myeloid leukemia cells by regulating miR-5195-3p/GRB10 axis. Hematol. Oncol. 2021, 39, 390–400. [Google Scholar] [CrossRef] [PubMed]
- Bi, J.; Pu, Y.; Yu, X. Exosomal circ_0004136 enhances the progression of pediatric acute myeloid leukemia depending on the regulation of miR-570-3p/TSPAN3 axis. Anti-Cancer Drugs 2021, 32, 802–811. [Google Scholar] [CrossRef]
- Kumar, B.; Garcia, M.; Weng, L.; Jung, X.; Murakami, J.L.; Hu, X.; McDonald, T.; Lin, A.; Kumar, A.R.; DiGiusto, D.L.; et al. Acute myeloid leukemia transforms the bone marrow niche into a leukemia-permissive microenvironment through exosome secretion. Leukemia 2018, 32, 575–587. [Google Scholar] [CrossRef]
- Doebele, C.; Bonauer, A.; Fischer, A.; Scholz, A.; Reiss, Y.; Urbich, C.; Hofmann, W.-K.; Zeiher, A.M.; Dimmeler, S. Members of the microRNA-17-92 cluster exhibit a cell-intrinsic antiangiogenic function in endothelial cells. Blood 2010, 115, 4944–4950. [Google Scholar] [CrossRef]
- Jafarzadeh, N.; Gholampour, M.A.; Alivand, M.R.; Kavousi, S.; Arzi, L.; Rad, F.; Sadeghizadeh, M.; Pornour, M. CML derived exosomes promote tumor favorable functional performance in T cells. BMC Cancer 2021, 21, 1002. [Google Scholar] [CrossRef]
- Zhang, H.-M.; Li, Q.; Zhu, X.; Liu, W.; Hu, H.; Liu, T.; Cheng, F.-J.; You, Y.; Zhong, Z.; Zou, P.; et al. miR-146b-5p within BCR-ABL1–Positive Microvesicles Promotes Leukemic Transformation of Hematopoietic Cells. Cancer Res. 2016, 76, 2901–2911. [Google Scholar] [CrossRef]
- Li, S. Src-family kinases in the development and therapy of Philadelphia chromosome-positive chronic myeloid leukemia and acute lymphoblastic leukemia. Leuk. Lymphoma 2008, 49, 19–26. [Google Scholar] [CrossRef]
- Tadokoro, H.; Umezu, T.; Ohyashiki, K.; Hirano, T.; Ohyashiki, J.H. Exosomes derived from hypoxic leukemia cells enhance tube formation in endothelial cells. J. Biol. Chem. 2013, 288, 34343–34351. [Google Scholar] [CrossRef] [Green Version]
- Taverna, S.; Flugy, A.; Saieva, L.; Kohn, E.C.; Santoro, A.; Meraviglia, S.; De Leo, G.; Alessandro, R. Role of exosomes released by chronic myelogenous leukemia cells in angiogenesis. Int. J. Cancer 2012, 130, 2033–2043. [Google Scholar] [CrossRef] [PubMed]
- Forte, D.; Barone, M.; Morsiani, C.; Simonetti, G.; Fabbri, F.; Bruno, S.; Bandini, E.; Sollazzo, D.; Collura, S.; Deregibus, M.C.; et al. Distinct profile of CD34(+) cells and plasma-derived extracellular vesicles from triple-negative patients with Myelofibrosis reveals potential markers of aggressive disease. J. Exp. Clin. Cancer Res. 2021, 40, 49. [Google Scholar] [CrossRef] [PubMed]
- Yan, W.; Song, L.; Wang, H.; Hu, L.; Yang, Y. Extracellular vesicles carrying miRNA-181b-5p affects the malignant progression of acute lymphoblastic leukemia. J. Transl. Med. 2021, 19, 511. [Google Scholar] [CrossRef] [PubMed]
- Haque, S.; Vaiselbuh, S.R. Silencing of Exosomal miR-181a Reverses Pediatric Acute Lymphocytic Leukemia Cell Proliferation. Pharmaceuticals 2020, 13, 241. [Google Scholar] [CrossRef]
- Smallwood, D.T.; Apollonio, B.; Willimott, S.; Lezina, L.; Alharthi, A.; Ambrose, A.R.; de Rossi, G.; Ramsay, A.G.; Wagner, S.D. Extracellular vesicles released by CD40/IL-4-stimulated CLL cells confer altered functional properties to CD4+ T cells. Blood 2016, 128, 542–552. [Google Scholar] [CrossRef]
- Dörsam, B.; Bösl, T.; Reiners, K.S.; Barnert, S.; Schubert, R.; Shatnyeva, O.; Zigrino, P.; Engert, A.; Hansen, H.P.; Von Strandmann, E.P. Hodgkin Lymphoma-Derived Extracellular Vesicles Change the Secretome of Fibroblasts Toward a CAF Phenotype. Front. Immunol. 2018, 9, 1358. [Google Scholar] [CrossRef]
- Provencio, M.; Rodríguez, M.; Cantos, B.; Sabín, P.; Quero, C.; García-Arroyo, F.R.; Rueda, A.; Maximiano, C.; Rodríguez-Abreu, D.; Sánchez, A.; et al. mRNA in exosomas as a liquid biopsy in non-Hodgkin Lymphoma: A multicentric study by the Spanish Lymphoma Oncology Group. Oncotarget 2017, 8, 50949–50957. [Google Scholar] [CrossRef]
- Wang, J.; De Veirman, K.; Faict, S.; Frassanito, M.A.; Ribatti, D.; Vacca, A.; Menu, E. Multiple myeloma exosomes establish a favourable bone marrow microenvironment with enhanced angiogenesis and immunosuppression. J. Pathol. 2016, 239, 162–173. [Google Scholar] [CrossRef]
- Wang, J.; De Veirman, K.; De Beule, N.; Maes, K.; De Bruyne, E.; Van Valckenborgh, E.; Vanderkerken, K.; Menu, E. The bone marrow microenvironment enhances multiple myeloma progression by exosome-mediated activation of myeloid-derived suppressor cells. Oncotarget 2015, 6, 43992–44004. [Google Scholar] [CrossRef]
- Umezu, T.; Tadokoro, H.; Azuma, K.; Yoshizawa, S.; Ohyashiki, K.; Ohyashiki, J.H. Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances angiogenesis by targeting factor-inhibiting HIF-1. Blood 2014, 124, 3748–3757. [Google Scholar] [CrossRef] [Green Version]
- Li, B.; Xu, H.; Han, H.; Song, S.; Zhang, X.; Ouyang, L.; Qian, C.; Hong, Y.; Qiu, Y.; Zhou, W.; et al. Exosome-mediated transfer of lncRUNX2-AS1 from multiple myeloma cells to MSCs contributes to osteogenesis. Oncogene 2018, 37, 5508–5519. [Google Scholar] [CrossRef] [PubMed]
- Raimondo, S.; Saieva, L.; Vicario, E.; Pucci, M.; Toscani, D.; Manno, M.; Raccosta, S.; Giuliani, N.; Alessandro, R.; Raccosta, S. Multiple myeloma-derived exosomes are enriched of amphiregulin (AREG) and activate the epidermal growth factor pathway in the bone microenvironment leading to osteoclastogenesis. J. Hematol. Oncol. 2019, 12, 2. [Google Scholar] [CrossRef] [PubMed]
- Raimondi, L.; De Luca, A.; Amodio, N.; Manno, M.; Raccosta, S.; Taverna, S.; Bellavia, D.; Naselli, F.; Fontana, S.; Schillaci, O.; et al. Involvement of multiple myeloma cell-derived exosomes in osteoclast differentiation. Oncotarget 2015, 6, 13772–13789. [Google Scholar] [CrossRef]
- Viola, S.; Traer, E.; Huan, J.; Hornick, N.I.; Tyner, J.W.; Agarwal, A.; Loriaux, M.M.; Johnstone, B.; Kurre, P. Alterations in acute myeloid leukaemia bone marrow stromal cell exosome content coincide with gains in tyrosine kinase inhibitor resistance. Br. J. Haematol. 2016, 172, 983–986. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Luo, X.-Q.; Feng, D.-D.; Zhang, X.-J.; Wu, J.; Zheng, Y.-S.; Chen, X.; Xu, L.; Chen, Y.-Q. Upregulation of microRNA-125b contributes to leukemogenesis and increases drug resistance in pediatric acute promyelocytic leukemia. Mol. Cancer 2011, 10, 108. [Google Scholar] [CrossRef] [PubMed]
- Bouvy, C.; Wannez, A.; Laloy, J.; Chatelain, C.; Dogné, J.-M. Transfer of multidrug resistance among acute myeloid leukemia cells via extracellular vesicles and their microRNA cargo. Leuk. Res. 2017, 62, 70–76. [Google Scholar] [CrossRef] [PubMed]
- Fei, F.; Joo, E.J.; Tarighat, S.S.; Schiffer, I.; Paz, H.; Fabbri, M.; Abdel-Azim, H.; Groffen, J.; Heisterkamp, N. B-cell precursor acute lymphoblastic leukemia and stromal cells communicate through Galectin-3. Oncotarget 2015, 6, 11378–11394. [Google Scholar] [CrossRef] [PubMed]
- van Eijndhoven, M.A.; Zijlstra, J.M.; Groenewegen, N.J.; Drees, E.E.; van Niele, S.; Baglio, S.R.; Koppers-Lalic, D.; van der Voorn, H.; Libregts, S.F.W.M.; Wauben, M.H.M.; et al. Plasma vesicle miRNAs for therapy response monitoring in Hodgkin lymphoma patients. JCI Insight 2016, 1, e89631. [Google Scholar] [CrossRef]
- Jiang, L.; Deng, T.; Wang, D.; Xiao, Y. Elevated Serum Exosomal miR-125b Level as a Potential Marker for Poor Prognosis in Intermediate-Risk Acute Myeloid Leukemia. Acta Haematol. 2018, 140, 183–192. [Google Scholar] [CrossRef]
- Chen, L.; Guo, Z.; Zhou, Y.; Ni, J.; Zhu, J.; Fan, X.; Chen, X.; Liu, Y.; Li, Z.; Zhou, H. microRNA-1246-containing extracellular vesicles from acute myeloid leukemia cells promote the survival of leukemia stem cells via the LRIG1-meditated STAT3 pathway. Aging 2021, 13, 13644–13662. [Google Scholar] [CrossRef]
- Bernardi, S.; Farina, M. Exosomes and Extracellular Vesicles in Myeloid Neoplasia: The Multiple and Complex Roles Played by These “Magic Bullets”. Biology 2021, 10, 105. [Google Scholar] [CrossRef] [PubMed]
- Kontopoulou, E.; Strachan, S.; Reinhardt, K.; Kunz, F.; Walter, C.; Walkenfort, B.; Jastrow, H.; Hasenberg, M.; Giebel, B.; von Neuhoff, N.; et al. Evaluation of dsDNA from extracellular vesicles (EVs) in pediatric AML diagnostics. Ann. Hematol. 2020, 99, 459–475. [Google Scholar] [CrossRef]
- Cai, J.; Han, Y.; Ren, H.; Chen, C.; He, D.; Zhou, L.; Eisner, G.M.; Asico, L.D.; Jose, P.A.; Zeng, C. Extracellular vesicle-mediated transfer of donor genomic DNA to recipient cells is a novel mechanism for genetic influence between cells. J. Mol. Cell Biol. 2013, 5, 227–238. [Google Scholar] [CrossRef] [PubMed]
- Cai, J.; Wu, G.; Tan, X.; Han, Y.; Chen, C.; Li, C.; Wang, N.; Zou, X.; Chen, X.; Zhou, F.; et al. Transferred BCR/ABL DNA from K562 Extracellular Vesicles Causes Chronic Myeloid Leukemia in Immunodeficient Mice. PLoS ONE 2014, 9, e105200. [Google Scholar]
- Jurj, A.; Pasca, S.; Teodorescu, P.; Tomuleasa, C.; Berindan-Neagoe, I. Basic knowledge on BCR-ABL1-positive extracellular vesicles. Biomark. Med. 2020, 14, 451–458. [Google Scholar] [CrossRef]
- Caivano, A.; Laurenzana, I.; De Luca, L.; La Rocca, F.; Simeon, V.; Trino, S.; D’Auria, F.; Traficante, A.; Maietti, M.; Izzo, T.; et al. High serum levels of extracellular vesicles expressing malignancy-related markers are released in patients with various types of hematological neoplastic disorders. Tumor Biol. 2015, 36, 9739–9752. [Google Scholar] [CrossRef]
- Taniguchi, Y.; Tanaka, H.; Luis, E.J.; Sakai, K.; Kumode, T.; Sano, K.; Serizawa, K.; Rai, S.; Morita, Y.; Hanamoto, H.; et al. Elevated plasma levels of procoagulant microparticles are a novel risk factor for thrombosis in patients with myeloproliferative neoplasms. Int. J. Hematol. 2017, 106, 691–703. [Google Scholar] [CrossRef]
- Trappenburg, M.C.; Van Schilfgaarde, M.; Marchetti, M.; Spronk, H.M.; ten Cate, H.; Leyte, A.; Terpstra, W.E.; Falanga, A. Elevated procoagulant microparticles expressing endothelial and platelet markers in essential thrombocythemia. Haematologica 2009, 94, 911–918. [Google Scholar] [CrossRef]
- Li, J.-W.; Shi, D.; Wan, X.-C.; Hu, J.; Su, Y.-F.; Zeng, Y.-P.; Hu, Z.-J.; Yu, B.-H.; Zhang, Q.-L.; Wei, P.; et al. Universal extracellular vesicles and PD-L1+ extracellular vesicles detected by single molecule array technology as circulating biomarkers for diffuse large B cell lymphoma. OncoImmunology 2021, 10, 1995166. [Google Scholar] [CrossRef]
- Manier, S.; Liu, C.-J.; Avet-Loiseau, H.; Park, J.; Shi, J.; Campigotto, F.; Salem, K.Z.; Huynh, D.; Glavey, S.V.; Rivotto, B.; et al. Prognostic role of circulating exosomal miRNAs in multiple myeloma. Blood 2017, 129, 2429–2436. [Google Scholar] [CrossRef]
- Wojtuszkiewicz, A.; Schuurhuis, G.J.; Kessler, F.L.; Piersma, S.R.; Knol, J.C.; Pham, T.V.; Jansen, G.; Musters, R.J.P.; van Meerloo, J.; Assaraf, Y.G.; et al. Exosomes Secreted by Apoptosis-Resistant Acute Myeloid Leukemia (AML) Blasts Harbor Regulatory Network Proteins Potentially Involved in Antagonism of Apoptosis. Mol. Cell. Proteom. 2016, 15, 1281–1298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gärtner, K.; Luckner, M.; Wanner, G.; Zeidler, R. Engineering extracellular vesicles as novel treatment options: Exploiting herpesviral immunity in CLL. J. Extracell. Vesicles 2019, 8, 1573051. [Google Scholar] [CrossRef] [PubMed]
- Chen, Z.; You, L.; Wang, L.; Huang, X.; Liu, H.; Wei, J.Y.; Zhu, L.; Qian, W. Dual effect of DLBCL-derived EXOs in lymphoma to improve DC vaccine efficacy in vitro while favor tumorgenesis in vivo. J. Exp. Clin. Cancer Res. 2018, 37, 190. [Google Scholar] [CrossRef] [PubMed]
- Borrelli, C.; Ricci, B.; Vulpis, E.; Fionda, C.; Ricciardi, M.R.; Petrucci, M.T.; Masuelli, L.; Peri, A.; Cippitelli, M.; Zingoni, A.; et al. Drug-Induced Senescent Multiple Myeloma Cells Elicit NK Cell Proliferation by Direct or Exosome-Mediated IL15 Trans-Presentation. Cancer Immunol. Res. 2018, 6, 860–869. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Hendrix, A.; Hernot, S.; Lemaire, M.; De Bruyne, E.; Van Valckenborgh, E.; Lahoutte, T.; De Wever, O.; Vanderkerken, K.; Menu, E. Bone marrow stromal cell–derived exosomes as communicators in drug resistance in multiple myeloma cells. Blood 2014, 124, 555–566. [Google Scholar] [CrossRef]
- Arber, D.A.; Orazi, A.; Hasserjian, R.; Thiele, J.; Borowitz, M.J.; Le Beau, M.M.; Bloomfield, C.D.; Cazzola, M.; Vardiman, J.W. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 2016, 127, 2391–2405. [Google Scholar] [CrossRef]
- Greenberg, P.L.; Tuechler, H.; Schanz, J.; Sanz, G.; Garcia-Manero, G.; Solé, F.; Bennett, J.M.; Bowen, D.; Fenaux, P.; Dreyfus, F.; et al. Revised International Prognostic Scoring System for Myelodysplastic Syndromes. Blood 2012, 120, 2454–2465. [Google Scholar] [CrossRef]
- Zuo, Z.; Calin, G.; De Paula, H.M.; Medeiros, L.J.; Fernandez, M.H.; Shimizu, M.; Garcia-Manero, G.; Bueso-Ramos, C.E. Circulating microRNAs let-7a and miR-16 predict progression-free survival and overall survival in patients with myelodysplastic syndrome. Blood 2011, 118, 413–415. [Google Scholar] [CrossRef]
- Kim, Y.; Cheong, J.; Kim, Y.; Eom, J.; Jeung, H.; Kim, S.; Hwang, D.; Kim, J.S.; Kim, H.J.; Min, Y.H. Serum microRNA-21 as a potential biomarker for response to hypomethylating agents in myelodysplastic syndromes. PLoS ONE 2014, 9, e86933. [Google Scholar] [CrossRef]
- Enjeti, A.K.; Ariyarajah, A.; D’Crus, A.; Riveros, C.; Seldon, M.; Lincz, L.F. Circulating microvesicles are less procoagulant and carry different miRNA cargo in myelodysplasia. Blood Cells Mol. Dis. 2019, 74, 37–43. [Google Scholar] [CrossRef]
- Skog, J.; Würdinger, T.; Van Rijn, S.; Meijer, D.H.; Gainche, L.; Curry, W.T., Jr.; Carter, B.S.; Krichevsky, A.M.; Breakefield, X.O. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat. Cell Biol. 2008, 10, 1470–1476. [Google Scholar] [CrossRef] [PubMed]
- Bi, L.; Sun, L.; Jin, Z.; Zhang, S.; Shen, Z. MicroRNA-10a/b are regulators of myeloid differentiation and acute myeloid leukemia. Oncol. Lett. 2018, 15, 5611–5619. [Google Scholar] [CrossRef] [PubMed]
- Huan, J.; Hornick, N.I.; Shurtleff, M.J.; Skinner, A.M.; Goloviznina, N.A.; Roberts, C.T.; Kurre, P. RNA Trafficking by Acute Myelogenous Leukemia Exosomes. Cancer Res. 2013, 73, 918–929. [Google Scholar] [CrossRef] [PubMed]
- Vargas Romero, P.; Cialfi, S.; Palermo, R.; De Blasio, C.; Checquolo, S.; Bellavia, D.; Chiaretti, S.; Foà, R.; Amadori, A.; Gulino, A.; et al. The deregulated expression of miR-125b in acute myeloid leukemia is dependent on the transcription factor C/EBPalpha. Leukemia 2015, 29, 2442–2445. [Google Scholar] [CrossRef]
- Battula, V.L.; Le, P.M.; Sun, J.C.; Nguyen, K.; Yuan, B.; Zhou, X.; Sonnylal, S.; McQueen, T.; Ruvolo, V.; Michel, K.A.; et al. AML-induced osteogenic differentiation in mesenchymal stromal cells supports leukemia growth. JCI Insight 2017, 2, e90036. [Google Scholar] [CrossRef]
- Fang, Y.; Garnier, D.; Lee, T.H.; D’Asti, E.; Montermini, L.; Meehan, B.; Rak, J. PML–RARa modulates the vascular signature of extracellular vesicles released by acute promyelocytic leukemia cells. Angiogenesis 2016, 19, 25–38. [Google Scholar] [CrossRef]
- Park, J.E.; Tan, H.S.; Datta, A.; Lai, R.C.; Zhang, H.; Meng, W.; Lim, S.K.; Sze, S.K. Hypoxic Tumor Cell Modulates Its Microenvironment to Enhance Angiogenic and Metastatic Potential by Secretion of Proteins and Exosomes. Mol. Cell. Proteom. 2010, 9, 1085–1099. [Google Scholar] [CrossRef]
- Hong, C.-S.; Sharma, P.; Yerneni, S.S.; Simms, P.; Jackson, E.K.; Whiteside, T.L.; Boyiadzis, M. Circulating exosomes carrying an immunosuppressive cargo interfere with cellular immunotherapy in acute myeloid leukemia. Sci. Rep. 2017, 7, 14684. [Google Scholar] [CrossRef]
- Izadirad, M.; Huang, Z.; Jafari, F.; Hamidieh, A.A.; Gharehbaghian, A.; Li, Y.-D.; Jafari, L.; Chen, Z.-S. Extracellular Vesicles in Acute Leukemia: A Mesmerizing Journey with a Focus on Transferred microRNAs. Front. Cell Dev. Biol. 2021, 9, 766371. [Google Scholar] [CrossRef]
- Kang, K.-W.; Jung, J.-H.; Hur, W.; Park, J.; Shin, H.; Choi, B.; Jeong, H.; Kim, D.S.; Yu, E.S.; Lee, S.R.; et al. The Potential of Exosomes Derived from Chronic Myelogenous Leukaemia Cells as a Biomarker. Anticancer Res. 2018, 38, 3935–3942. [Google Scholar] [CrossRef]
- Bernardi, S.; Foroni, C.; Zanaglio, C.; Re, F.; Polverelli, N.; Turra, A.; Morello, E.; Farina, M.; Cattina, F.; Gandolfi, L.; et al. Feasibility of tumor-derived exosome enrichment in the onco-hematology leukemic model of chronic myeloid leukemia. Int. J. Mol. Med. 2019, 44, 2133–2144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Catani, L.; Cavo, M.; Palandri, F. The Power of Extracellular Vesicles in Myeloproliferative Neoplasms: “Crafting” a Microenvironment That Matters. Cells 2021, 10, 2316. [Google Scholar] [CrossRef] [PubMed]
- Aswad, M.H.; Kissova, J.; Ovesna, P.; Rihova, L.; Penka, M. The Clinical Significance of Circulating Microparticles Concerning Thrombosis in BCR/ABL1-negative Myeloproliferative Neoplasms. In Vivo 2021, 35, 3345–3353. [Google Scholar] [CrossRef] [PubMed]
- Ahadon, M.; Aziz, S.A.; Wong, C.L.; Leong, C.F. Plasma-derived microparticles in polycythaemia vera. Malays. J. Pathol. 2018, 40, 41–48. [Google Scholar] [PubMed]
- Poisson, J.; Tanguy, M.; Davy, H.; Camara, F.; El Mdawar, M.-B.; Kheloufi, M.; Dagher, T.; Devue, C.; Lasselin, J.; Plessier, A.; et al. Erythrocyte-derived microvesicles induce arterial spasms in JAK2V617F myeloproliferative neoplasm. J. Clin. Investig. 2020, 130, 2630–2643. [Google Scholar] [CrossRef]
- Charpentier, A.; Lebreton, A.; Rauch, A.; Bauters, A.; Trillot, N.; Nibourel, O.; Tintillier, V.; Wemeau, M.; Demory, J.-L.; Preudhomme, C.; et al. Microparticle phenotypes are associated with driver mutations and distinct thrombotic risks in essential thrombocythemia. Haematologica 2016, 101, e365–e368. [Google Scholar] [CrossRef]
- Piccin, A.; Steurer, M.; Feistritzer, C.; Murphy, C.; Eakins, E.; Van Schilfgaarde, M.; Corvetta, D.; Di Pierro, A.M.; Pusceddu, I.; Marcheselli, L.; et al. Observational retrospective study of vascular modulator changes during treatment in essential thrombocythemia. Transl. Res. 2017, 184, 21–34. [Google Scholar] [CrossRef]
- Barone, M.; Ricci, F.; Sollazzo, D.; Ottaviani, E.; Romano, M.; Auteri, G.; Bartoletti, D.; Reggiani, M.L.B.; Vianelli, N.; Tazzari, P.L.; et al. Circulating megakaryocyte and platelet microvesicles correlate with response to ruxolitinib and distinct disease severity in patients with myelofibrosis. Br. J. Haematol. 2019, 185, 987–991. [Google Scholar] [CrossRef]
- Patel, S.J.; Darie, C.C.; Clarkson, B.D. Exosome mediated growth effect on the non-growing pre-B acute lymphoblastic leukemia cells at low starting cell density. Am. J. Transl. Res. 2016, 8, 3614–3629. [Google Scholar]
- Egyed, B.; Kutszegi, N.; Sági, J.C.; Gézsi, A.; Rzepiel, A.; Visnovitz, T.; Lőrincz, P.; Müller, J.; Zombori, M.; Szalai, C.; et al. MicroRNA-181a as novel liquid biopsy marker of central nervous system involvement in pediatric acute lymphoblastic leukemia. J. Transl. Med. 2020, 18, 250. [Google Scholar] [CrossRef]
- Fei, F.; Abdel-Azim, H.; Lim, M.; Arutyunyan, A.; Von Itzstein, M.; Groffen, J.; Heisterkamp, N. Galectin-3 in pre-B acute lymphoblastic leukemia. Leukemia 2013, 27, 2385–2388. [Google Scholar] [CrossRef] [PubMed]
- Crompot, E.; Van Damme, M.; Pieters, K.; Vermeersch, M.; Perez-Morga, D.; Mineur, P.; Maerevoet, M.; Meuleman, N.; Bron, D.; Lagneaux, L.; et al. Extracellular vesicles of bone marrow stromal cells rescue chronic lymphocytic leukemia B cells from apoptosis, enhance their migration and induce gene expression modifications. Haematologica 2017, 102, 1594–1604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aguilar-Hernandez, M.M.; Camacho, J.C.R.; Garcia, G.G. Extracellular vesicles and their associated miRNAs as potential prognostic biomarkers in chronic lymphocytic leukemia. Curr. Oncol. Rep. 2021, 23, 66. [Google Scholar] [CrossRef]
- Cox, M.J.; Lucien, F.; Sakemura, R.; Boysen, J.C.; Kim, Y.; Horvei, P.; Roman, C.M.; Hansen, M.J.; Tapper, E.E.; Siegler, E.L.; et al. Leukemic extracellular vesicles induce chimeric antigen receptor T cell dysfunction in chronic lymphocytic leukemia. Mol. Ther. 2021, 29, 1529–1540. [Google Scholar] [CrossRef] [PubMed]
- Navarro-Tableros, V.; Gomez, Y.; Camussi, G.; Brizzi, M.F. Extracellular Vesicles: New Players in Lymphomas. Int. J. Mol. Sci. 2018, 20, 41. [Google Scholar] [CrossRef]
- Koch, R.; Demant, M.; Aung, T.; Diering, N.; Cicholas, A.; Chapuy, B.; Wenzel, D.; Lahmann, M.; Güntsch, A.; Kiecke, C.; et al. Populational equilibrium through exosome-mediated Wnt signaling in tumor progression of diffuse large B-cell lymphoma. Blood 2014, 123, 2189–2198. [Google Scholar] [CrossRef]
- Carvalho, A.S.; Baeta, H.; Henriques, A.F.A.; Ejtehadifar, M.; Tranfield, E.M.; Sousa, A.L.; Farinho, A.; Silva, B.C.; Cabeçadas, J.; Gameiro, P.; et al. Proteomic Landscape of Extracellular Vesicles for Diffuse Large B-Cell Lymphoma Subtyping. Int. J. Mol. Sci. 2021, 22, 11004. [Google Scholar] [CrossRef]
- Feng, Y.; Zhong, M.; Zeng, S.; Wang, L.; Liu, P.; Xiao, X.; Liu, Y. Exosome-derived miRNAs as predictive biomarkers for diffuse large B-cell lymphoma chemotherapy resistance. Epigenomics 2019, 11, 35–51. [Google Scholar] [CrossRef]
- Wang, X.; He, L.; Huang, X.; Zhang, S.; Cao, W.; Che, F.; Zhu, Y.; Dai, J. Recent Progress of Exosomes in Multiple Myeloma: Pathogenesis, Diagnosis, Prognosis and Therapeutic Strategies. Cancers 2021, 13, 1635. [Google Scholar] [CrossRef]
- Iaccino, E.; Mimmi, S.; Dattilo, V.; Marino, F.; Candeloro, P.; Di Loria, A.; Marimpietri, D.; Pisano, A.; Albano, F.; Vecchio, E.; et al. Monitoring multiple myeloma by idiotype-specific peptide binders of tumor-derived exosomes. Mol. Cancer 2017, 16, 159. [Google Scholar] [CrossRef]
- Ayers, L.; Pink, R.; Carter, D.R.F.; Nieuwland, R. Clinical requirements for extracellular vesicle assays. J. Extracell. Vesicles 2019, 8, 1593755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
EV Role | Target | Effect | Haematological Malignancy | Ref. |
---|---|---|---|---|
Biomarker | Upregulated miR-126-3p, miR-125a-5p, miR-199a-3p, miR-151a-3p and miR-423-5p, | - | MDS | [7] |
Downregulated miR-16, miR-17, miR-20a, miR-21, miR-126, miR-181a, miR-146a, and miR-155 | - | MDS | [8] | |
Downregulated miR-28 | Increased bleeding | MDS | [9] | |
miR-10b | Suppressed myeloid differentiation and inhibited apoptosis | AML | [10] | |
miR-150 and miR-155 | Suppressed proliferative and differentiation capacity of HSPC | AML | [11] | |
miR-125b, miR-26a-5p | inhibited the Expression of proapoptotic molecules | AML | [12] | |
lnc-RNA, such as circ-0009910 and circ-0004136 | Promote cell cycle progression and AML cell viability and invasion | AML | [13,14] | |
DKK-1 | Inhibited osteogenesis | AML | [15] | |
IL-8, VEGF | Stimulated angiogenesis | AML | [16] | |
IL-10, IL-6, IL-17, NO, FOXP3 | Facilitated tumour survival | CML | [17] | |
Dysregulation of miR-146b-5p | Associated with leukaemia transformation | CML | [18] | |
miR-92a, miR-210, IL-8 | Promoted angiogenesis | CML | [19,20,21] | |
miR-34a-5p, -miR-127-3p, miR-212-3p, miR-361 | Promoted survival and migration of CD34+ cells | MF | [22] | |
Upregulation of miR-181b-5p | Promoted survival, migration and invasion of ALL cells | ALL | [23] | |
Upregulation of survival genes and downregulation of proapoptotic genes via miR-181a | Leukaemic proliferation and survival | ALL | [24] | |
miR-363 | Altered the function of CD4+ T cells | CLL | [25] | |
Promote TNF-a/NFkB signalling | Altered ME to support tumour growth and survival | HL | [26] | |
Elevated BCL-6 expression | - | DLBCL | [27] | |
Upregulation of I/S molecules (such as iNOS) | Promote MDSCs and create I/S ME | MM | [28,29] | |
miR-135b | Upregulation of HIF-1, which promoted angiogenesis | MM | [30] | |
IncRUNX2-AS1 | Osteogenesis suppression | MM | [31] | |
Increased AREG | Increased bone resorption | MM | [32] | |
Increased CXCR4, RANKL, CTSK, MMP9, TRAP | Promoted osteoclast migration, differentiation, and survival | MM | [33] | |
Drug resistance | TGF-β, miR-155, miR-375 | Stromal protection of AML cells | AML | [34] |
miR-125b | - | APML | [35] | |
MRP-1 | - | AML | [36] | |
Galectin-3 | Protective effect of stromal fibroblasts on ALL cells | ALL | [37] | |
Increased AKT mRNA | Reduced response to rituximab | FL | [27] | |
miR-99a-5p and miR-125b-5p | Chemotherapy resistance and poor outcomes | HL | [38] | |
Disease outcomes | Increased miR-125 | Aggressive course, higher risk of relapse and significantly shorter survival | AML | [39] |
Increased miR-10b | Significantly shorter OS and DFS | AML | [10] | |
Increased miR-1246, miR-532 and miR-125b | Predicted for relapse and poor outcomes | AML | [17,18,19,20,21,39,40,41,42,43,44,45,46] | |
Elevated TF | Thrombotic events | MPD | [47] | |
Elevated vWF and thrombin | Hypercoagulable state | ET | [48] | |
BCL-6 and c-myc mRNA | Predict shorter OS and worse PFS | FL | [27] | |
High levels of CD9/CD63 and PD-L1/CD63 EVs | Correlate with therapeutic failure and poor outcomes | DLBCL | [49] | |
let-7b and miR-18a | Predicted for worse OS and PFS | MM | [50] | |
Target | Antiapoptotic proteins (such as MCL-1, BCL-2 and BCL-XL) | Induction of apoptosis resistance | AML | [51] |
Silencing of miR-181a | Inhibition of leukaemic cell proliferation | ALL | [24] | |
EVs carrying gp350, CD40 and pp65 | Exhibited strong Ag-presenting capacity and induces specific cytotoxic T cells | CLL | [52] | |
Exchanged Wnt signals between SP and non-SP cells | Allowed tumour cell communication and progression | HL | [26] | |
Vaccine using TEX-carrying TAA | Enhanced antitumour responses in mouse models | Lymphoma | [53] | |
Inhibition of EV formation | Amelioration of lytic lesions in mice | MM | [31] | |
Melphalan- and doxorubicin-induced EVs | Carried IL15/IL15RA, which promotes an antitumour response | MM | [54] | |
MSC-derived EVs | Reversed the bortezomib-induced bcl-2 inhibition and inhibit the cleavage of cas3, cas9 and PARP | MM | [55] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the author. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Lazana, I. Extracellular Vesicles in Haematological Disorders: A Friend or a Foe? Int. J. Mol. Sci. 2022, 23, 10118. https://doi.org/10.3390/ijms231710118
Lazana I. Extracellular Vesicles in Haematological Disorders: A Friend or a Foe? International Journal of Molecular Sciences. 2022; 23(17):10118. https://doi.org/10.3390/ijms231710118
Chicago/Turabian StyleLazana, Ioanna. 2022. "Extracellular Vesicles in Haematological Disorders: A Friend or a Foe?" International Journal of Molecular Sciences 23, no. 17: 10118. https://doi.org/10.3390/ijms231710118
APA StyleLazana, I. (2022). Extracellular Vesicles in Haematological Disorders: A Friend or a Foe? International Journal of Molecular Sciences, 23(17), 10118. https://doi.org/10.3390/ijms231710118