• No results found

3.2.8.2 – Lysate of exosomes with NP-40 lysis buffer Reagents:

6 Future perspectives

In this thesis we have established a protocol for exosome isolation from plasma using size-exclusion chromatography. With a combination of mass spectrometric analysis and electron microscopy, we were able to define fractions 7-8 as containing exosomes with least plasma protein contamination. These fractions also contained relative homogenously sized vesicles within the size-range of exosomes, and selective expression of two acknowledged exosomal markers. In this thesis we did not detect the exosomal marker CD63 by western blotting, an option might be to use LGALS3BP and CD5L instead. Also, the size of the isolated exosomes should be quantified by the use of the nanosight technology.

Further studies should include MS analysis of more control samples, as only exosomes from 2 healthy controls were included in this study. Also, exosomes from AML and T-ALL patients should be investigated by MS and compared to the results from pre-B ALL patients.

Moreover, another attempt at isolating blast-specific exosomes should be made, as we never attempted their separation from the SEC fractions. MS analysis of blast-enriched exosomes could yield detection of low abundance proteins, e.g. ligands for activating receptors, which could be below threshold for detection in a heterogeneous population as investigated here.

Also a protocol to perform immunophenotyping of the exosomes by flow cytometry could be developed. There exists protocols to capture exosomes on latex beads, which allows staining exosomes with antibodies for flow cytometric analysis. The advantage with this approach is the high sensitivity of antibodies that could lead to identification of low abundance proteins not detected by MS.

It would be interesting to test whether exosomes derived from patients have suppressive effects on NK cells. This could be achieved by culturing PBMC with exosomes, and analyze functional activity and expression of activating NK cell receptors by flow cytometry.

We isolated exosomes for future miRNA/RNA analysis, and a future directive could be to investigate whether the exosomes contain RNA for activating ligands or miRNA species that may influence gene expression of activating receptors in NK cells.

In terms of analysis of expression of activating ligands on leukemic cells, more samples should be included. In a follow-up study, also B7-H6, the other ligand for NKp30, and CD112 for DNAM-1 should be included. Also, it could be desirable to include flow

cytometric analysis of the ligands, as mRNA expression levels not necessarily predict protein expression.

References

1. Olivieri, A., et al., Imatinib for refractory chronic graft-versus-host disease with fibrotic features. Blood, 2009. 114(3): p. 709-18.

2. Patt, H.M., et al., Cysteine Protection against X Irradiation. Science, 1949.

110(2852): p. 213-4.

3. Owen, J.A., J. Punt, and S.A. Stranford, Kuby Immunology. 2009.

4. Abt, M.C., et al., Commensal bacteria calibrate the activation threshold of innate antiviral immunity. Immunity, 2012. 37(1): p. 158-70.

5. Whalen, M.M., et al., Expression of CD16, CD18 and CD56 in tributyltin-exposed human natural killer cells. ELSEVIER, 2002: p. 159-176.

6. Mavilio, D., et al., Characterization of CD56-/CD16+ natural killer (NK) cells: a highly dysfunctional NK subset expanded in HIV-infected viremic individuals. Proc Natl Acad Sci U S A, 2005. 102(8): p. 2886-91.

7. Kruse, V., et al., Human Induced Pluripotent Stem Cells Are Targets for Allogeneic and Autologous Natural Killer (NK) Cells and Killing Is Partly Mediated by the Activating NK Receptor DNAM-1. PLoS One, 2015. 10(5): p. e0125544.

8. Kulkarni, A.G., R.S. Paranjape, and M.R. Thakar, Higher Expression of Activating Receptors on Cytotoxic NK Cells is Associated with Early Control on HIV-1C Multiplication. Front Immunol, 2014. 5: p. 222.

9. Bauer, S., et al., Activation of NK Cells and T Cells by NKG2D, a Receptor for Stress-Inducible MICA SCIENCE, 1999. 285: p. 227-229.

10. Allan, A.J., et al., Cattle NK Cell Heterogeneity and the Influence of MHC Class I. J p38 Kinase Pathway. The Journal of Immunology, 2000. 164(12): p. 6244-6251.

13. Woodruff, M.F. and M. Sparrow, Further observation on the induction of tolerance of skin homografts in rats. Transplant Bull, 1957. 4(4): p. 157-9.

14. Liu, D., et al., Two modes of lytic granule fusion during degranulation by natural killer cells. Immunol Cell Biol, 2011. 89(6): p. 728-38.

15. Cooke, K.R., et al., Tumor necrosis factor- alpha production to lipopolysaccharide stimulation by donor cells predicts the severity of experimental acute graft-versus-host disease. J Clin Invest, 1998. 102(10): p. 1882-91.

16. Dang, V.T., et al., Fasting enhances TRAIL-mediated liver natural killer cell activity via HSP70 upregulation. PLoS One, 2014. 9(10): p. e110748.

17. Maksimovic-Ivanic, D., et al., Resistance to TRAIL and how to surmount it.

Immunol Res, 2012. 52(1-2): p. 157-68.

18. Boak, J.L., M. Fox, and R.E. Wilson, Activity of lymphoid tissues from antilymphocyte-serum-treated mice. Lancet, 1967. 1(7493): p. 750-2.

19. Sagiv, A., et al., Granule exocytosis mediates immune surveillance of senescent cells.

Oncogene, 2013. 32(15): p. 1971-7.

20. Liu, Q., et al., Matrix metalloprotease inhibitors restore impaired NK cell-mediated antibody-dependent cellular cytotoxicity in human immunodeficiency virus type 1

65

21. Lo Nigro, C., et al., Evaluation of antibody-dependent cell-mediated cytotoxicity activity and cetuximab response in KRAS wild-type metastatic colorectal cancer patients. World J Gastrointest Oncol, 2016. 8(2): p. 222-30.

22. Rodriguez, J.M., et al., PyNTTTTGT and CpG immunostimulatory oligonucleotides:

effect on granulocyte/monocyte colony-stimulating factor (GM-CSF) secretion by human CD56+ (NK and NKT) cells. PLoS One, 2015. 10(2): p. e0117484.

23. Tourdot, B.E., et al., Immunoreceptor tyrosine-based inhibitory motif (ITIM)-mediated inhibitory signaling is regulated by sequential phosphorylation (ITIM)-mediated by distinct nonreceptor tyrosine kinases: a case study involving PECAM-1.

Biochemistry, 2013. 52(15): p. 2597-608.

24. Mahmood, S., et al., SHP-1 phosphatase is a critical regulator in preventing natural killer cell self-killing. PLoS One, 2012. 7(8): p. e44244.

25. Salih, H.R., et al., Functional expression and release of ligands for the activating immunoreceptor NKG2D in leukemia. Blood, 2003. 102(4): p. 1389-1396.

26. Bottino, C., et al., Identification of PVR (CD155) and Nectin-2 (CD112) as cell surface ligands for the human DNAM-1 (CD226) activating molecule. J Exp Med, 2003. 198(4): p. 557-67.

27. Park, Y.P., et al., Complex regulation of human NKG2D-DAP10 cell surface expression: opposing roles of the gammac cytokines and TGF-beta1. Blood, 2011.

118(11): p. 3019-27.

28. Mathe, G. and J. Bernard, [Trial therapy, by x-irradiation followed by the administration of homologous bone marrow cells, of highly-advanced spontaneous leukemia in AK mice]. Bull Assoc Fr Etud Cancer, 1958. 45(3): p. 289-300.

29. Wei, P., et al., Molecular dynamic simulation of the self-assembly of DAP12-NKG2C activating immunoreceptor complex. PLoS One, 2014. 9(8): p. e105560.

30. Moncrieffe, H., M. Coles, and B. Stockinger, The influence of CD4 T-cell subsets on control of CD4 T-cell-mediated graft-versus-host disease. Immunology, 2008.

125(4): p. 459-468.

31. Rayfield, L.S. and L. Brent, Tolerance, immunocompetence, and secondary disease in fully allogeneic radiation chimeras. Transplantation, 1983. 36(2): p. 183-9.

32. Siewiera, J., et al., Natural cytotoxicity receptor splice variants orchestrate the distinct functions of human natural killer cell subtypes. Nat Commun, 2015. 6: p.

10183.

33. Gilfillan, S., et al., DNAM-1 promotes activation of cytotoxic lymphocytes by nonprofessional antigen-presenting cells and tumors. J Exp Med, 2008. 205(13): p.

2965-73.

34. Nabekura, T., et al., Costimulatory molecule DNAM-1 is essential for optimal differentiation of memory natural killer cells during mouse cytomegalovirus infection. Immunity, 2014. 40(2): p. 225-34.

35. Thomas, E.D., et al., Marrow transplants in lethally irradiated dogs given methotrexate. Blood, 1962. 19: p. 217-28.

36. Harder, L., et al., Aberrant ZNF423 impedes B cell differentiation and is linked to adverse outcome of ETV6-RUNX1 negative B precursor acute lymphoblastic leukemia. J Exp Med, 2013. 210(11): p. 2289-304.

37. Koyama, D., et al., Proteasome inhibitors exert cytotoxicity and increase chemosensitivity via transcriptional repression of Notch1 in T-cell acute lymphoblastic leukemia. Leukemia, 2014. 28(6): p. 1216-26.

38. Thathia, S.H., et al., Epigenetic inactivation of TWIST2 in acute lymphoblastic leukemia modulates proliferation, cell survival and chemosensitivity.

Haematologica, 2012. 97(3): p. 371-8.

39. Meng, X., et al., GSI-I (Z-LLNle-CHO) inhibits gamma-secretase and the proteosome to trigger cell death in precursor-B acute lymphoblastic leukemia. Leukemia, 2011.

25(7): p. 1135-46.

40. Storb, R., R.H. Rudolph, and E.D. Thomas, Marrow grafts between canine siblings matched by serotyping and mixed leukocyte culture. J Clin Invest, 1971. 50(6): p.

1272-5.

41. Balgobind, B.V., et al., Evaluation of gene expression signatures predictive of cytogenetic and molecular subtypes of pediatric acute myeloid leukemia.

Haematologica, 2011. 96(2): p. 221-30.

42. Hamilton, B.L., M.J. Bevan, and R. Parkman, Anti-recipient cytotoxic T lymphocyte precursors are present in the spleens of mice with acute graft versus host disease due to minor histocompatibility antigens. J Immunol, 1981. 126(2): p. 621-5.

43. Korngold, R. and J. Sprent, Lethal graft-versus-host disease after bone marrow transplantation across minor histocompatibility barriers in mice. Prevention by removing mature T cells from marrow. J Exp Med, 1978. 148(6): p. 1687-98.

44. Baker, G.J., et al., Natural killer cells eradicate galectin-1-deficient glioma in the absence of adaptive immunity. Cancer Res, 2014. 74(18): p. 5079-90.

45. Swann, J.B. and M.J. Smyth, Immune surveillance of tumors. J Clin Invest, 2007.

117(5): p. 1137-46.

46. Berger, M., P.J. Wettstein, and R. Korngold, T cell subsets involved in lethal graft-versus-host disease directed to immunodominant minor histocompatibility antigens. Transplantation, 1994. 57(7): p. 1095-102.

47. Milane, L., et al., Exosome mediated communication within the tumor microenvironment. J Control Release, 2015. 219: p. 278-94.

48. Oksvold, M.P., et al., Expression of B-cell surface antigens in subpopulations of exosomes released from B-cell lymphoma cells. Clin Ther, 2014. 36(6): p. 847-862 e1.

49. Labani-Motlagh, A., et al., Differential expression of ligands for NKG2D and DNAM-1 receptors by epithelial ovarian cancer-derived exosomes and its influence on NK cell cytotoxicity. Tumour Biol, 2016. 37(4): p. 5455-66.

50. Hong, C.S., et al., Isolation of biologically active and morphologically intact exosomes from plasma of patients with cancer. J Extracell Vesicles, 2016. 5: p.

29289.

51. Cho, J.A., et al., Exosomes: a new delivery system for tumor antigens in cancer immunotherapy. Int J Cancer, 2005. 114(4): p. 613-22.

52. Pogge von Strandmann, E., et al., Human leukocyte antigen-B-associated transcript 3 is released from tumor cells and engages the NKp30 receptor on natural killer cells. Immunity, 2007. 27(6): p. 965-74.

53. Tan, S.S., et al., Therapeutic MSC exosomes are derived from lipid raft microdomains in the plasma membrane. J Extracell Vesicles, 2013. 2.

54. Zhang, B., et al., Immunotherapeutic potential of extracellular vesicles. Front Immunol, 2014. 5: p. 518.

55. Wolff, D., et al., Methotrexate-albumin and aminopterin-albumin effectively prevent experimental acute graft-versus-host disease. Transplantation, 2006.

82(4): p. 527-33.

67

56. Bellingham, S.A., et al., Exosomes: vehicles for the transfer of toxic proteins associated with neurodegenerative diseases? Front Physiol, 2012. 3: p. 124.

57. Ruutu, T., et al., Prophylaxis and treatment of GVHD: EBMT-ELN working group recommendations for a standardized practice. Bone Marrow Transplant, 2014.

49(2): p. 168-73.

58. Kahlert, C., et al., Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNA in the serum exosomes of patients with pancreatic cancer. J Biol Chem, 2014. 289(7): p. 3869-75.

59. Ohajekwe, O.A., M.A. Hardy, and S.F. Oluwole, Prevention of graft-versus-host disease and the induction of transplant tolerance by low-dose UV-B irradiation of BM cells combined with cyclosporine immunosuppression. Transplantation, 1995.

60(12): p. 1510-6.

60. Jakobsen, K.R., et al., Exosomal proteins as potential diagnostic markers in advanced non-small cell lung carcinoma. J Extracell Vesicles, 2015. 4: p. 26659.

61. Li, M., et al., Analysis of the RNA content of the exosomes derived from blood serum and urine and its potential as biomarkers. Philos Trans R Soc Lond B Biol Sci, 2014. 369(1652).

62. Chugh, P.E., et al., Systemically circulating viral and tumor-derived microRNAs in KSHV-associated malignancies. PLoS Pathog, 2013. 9(7): p. e1003484.

63. Iorio, M.V., et al., MicroRNA signatures in human ovarian cancer. Cancer Res, 2007. 67(18): p. 8699-707.

64. Lundholm, M., et al., Prostate tumor-derived exosomes down-regulate NKG2D expression on natural killer cells and CD8+ T cells: mechanism of immune evasion.

PLoS One, 2014. 9(9): p. e108925.

65. Hong, C.S., et al., Isolation and characterization of CD34+ blast-derived exosomes in acute myeloid leukemia. PLoS One, 2014. 9(8): p. e103310.

66. Ashiru, O., et al., Natural killer cell cytotoxicity is suppressed by exposure to the transplantation: a prospective randomized study of the additional effect of decontamination and laminar air flow isolation among patients receiving prophylactic systemic antibiotics. Scand J Infect Dis, 1987. 19(5): p. 559-67.

70. Fernandez-Messina, L., et al., Differential mechanisms of shedding of the glycosylphosphatidylinositol (GPI)-anchored NKG2D ligands. J Biol Chem, 2010.

285(12): p. 8543-51.

71. Deng, W., et al., Antitumor immunity. A shed NKG2D ligand that promotes natural killer cell activation and tumor rejection. Science, 2015. 348(6230): p. 136-139.

72. Boing, A.N., et al., Single-step isolation of extracellular vesicles by size-exclusion chromatography. J Extracell Vesicles, 2014. 3.

73. Lossos, I.S., et al., Optimization of quantitative real-time RT-PCR parameters for the study of lymphoid malignancies. Leukemia, 2003. 17(4): p. 789-95.

74. de Menezes-Neto, A., et al., Size-exclusion chromatography as a stand-alone methodology identifies novel markers in mass spectrometry analyses of plasma-derived vesicles from healthy individuals. J Extracell Vesicles, 2015. 4: p. 27378.

75. Murai, M., et al., Peyer's patch is the essential site in initiating murine acute and lethal graft-versus-host reaction. Nat Immunol, 2003. 4(2): p. 154-60.

76. Hill, G.R., Inflammation and bone marrow transplantation. Biol Blood Marrow Transplant, 2009. 15(1 Suppl): p. 139-41.

77. Seol, H.J., et al., Overexpression of CD99 Increases the Migration and Invasiveness of Human Malignant Glioma Cells. Genes Cancer, 2012. 3(9-10): p. 535-49.

78. Momen-Heravi, F., et al., Impact of biofluid viscosity on size and sedimentation efficiency of the isolated microvesicles. Front Physiol, 2012. 3: p. 162.

79. Baranyai, T., et al., Isolation of Exosomes from Blood Plasma: Qualitative and Quantitative Comparison of Ultracentrifugation and Size Exclusion Chromatography Methods. PLoS One, 2015. 10(12): p. e0145686.

80. Wysocki, C.A., et al., Leukocyte migration and graft-versus-host disease. Blood, 2005. 105(11): p. 4191-9.

81. Eisele, G., et al., TGF-beta and metalloproteinases differentially suppress NKG2D ligand surface expression on malignant glioma cells. Brain, 2006. 129(Pt 9): p.

2416-25.

82. Kato, N., et al., Regulation of the expression of MHC class I-related chain A, B (MICA, MICB) via chromatin remodeling and its impact on the susceptibility of leukemic cells to the cytotoxicity of NKG2D-expressing cells. Leukemia, 2007. 21(10): p.

2103-8.

83. Tsukagoshi, M., et al., Overexpression of natural killer group 2 member D ligands predicts favorable prognosis in cholangiocarcinoma. Cancer Sci, 2016. 107(2): p.

116-22.

84. McGilvray, R.W., et al., NKG2D ligand expression in human colorectal cancer reveals associations with prognosis and evidence for immunoediting. Clin Cancer Res, 2009. 15(22): p. 6993-7002.

85. de Kruijf, E.M., et al., NKG2D ligand tumor expression and association with clinical outcome in early breast cancer patients: an observational study. BMC Cancer, 2012. 12: p. 24.

86. Pende, D., et al., Analysis of the receptor-ligand interactions in the natural killer-mediated lysis of freshly isolated myeloid or lymphoblastic leukemias: evidence for the involvement of the Poliovirus receptor (CD155) and Nectin-2 (CD112). Blood, 2005. 105(5): p. 2066-2073.

87. Yu, Y., et al., Prevention of GVHD while sparing GVL effect by targeting Th1 and Th17 transcription factor T-bet and RORgammat in mice. Blood, 2011. 118(18): p.

5011-20.

88. Hatano, R., et al., Prevention of acute graft-versus-host disease by humanized anti-CD26 monoclonal antibody. Br J Haematol, 2013. 162(2): p. 263-77.

89. Chen, X.J., et al., B7-H6 protein expression has no prognostic significance in human gastric carcinoma. Pathol Oncol Res, 2014. 20(1): p. 203-7.

90. Clayton, A., et al., Human Tumor-Derived Exosomes Down-Modulate NKG2D Expression. The Journal of Immunology, 2008. 180(11): p. 7249-7258.

91. Clayton, A., et al., Human tumor-derived exosomes selectively impair lymphocyte responses to interleukin-2. Cancer Res, 2007. 67(15): p. 7458-66.

69

92. Calabretta, S., et al., Modulation of PKM alternative splicing by PTBP1 promotes gemcitabine resistance in pancreatic cancer cells. Oncogene, 2016. 35(16): p.

2031-9.

93. Burman, A.C., et al., IFNgamma differentially controls the development of idiopathic pneumonia syndrome and GVHD of the gastrointestinal tract. Blood, 2007. 110(3): p. 1064-72.

APPENDIX 1: PRIMERS