3,495
Views
36
CrossRef citations to date
0
Altmetric
Original Articles

Extracellular vesicles mediate low dose ionizing radiation-induced immune and inflammatory responses in the blood

, , , , , & show all
Pages 12-22 | Received 15 Sep 2017, Accepted 23 Feb 2018, Published online: 29 Mar 2018

References

  • Al-Mayah AH, Irons SL, Pink RC, Carter DR, Kadhim MA. 2012. Possible role of exosomes containing RNA in mediating nontargeted effect of ionizing radiation. Radiat Res. 177:539–545.
  • Albanese J, Dainiak N. 2000. Ionizing radiation alters Fas antigen ligand at the cell surface and on exfoliated plasma membrane-derived vesicles: implications for apoptosis and intercellular signaling. Radiat Res. 153:49–61.
  • Andaloussi EL, Mager I, Breakefield XO, Wood MJ. 2013. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 12:347–357.
  • Arai F, Suda T. 2007. Maintenance of quiescent hematopoietic stem cells in the osteoblastic niche. Ann N Y Acad Sci. 1106:41–53.
  • Ariyoshi K, Takabatake T, Shinagawa M, Kadono K, Daino K, Imaoka T, Kakinuma S, Nishimura M, Shimada Y. 2014. Age dependence of hematopoietic progenitor survival and chemokine family gene induction after gamma irradiation in bone marrow tissue in C3H/He mice. Radiat Res. 181:302–313.
  • Asimakopoulou A, Borkham-Kamphorst E, Tacke F, Weiskirchen R. 2016. Lipocalin-2 (NGAL/LCN2), a “help-me” signal in organ inflammation. Hepatology. 63:669–671.
  • Asimakopoulou A, Weiskirchen S, Weiskirchen R. 2016. Lipocalin 2 (LCN2) expression in hepatic malfunction and therapy. Front Physiol. 7:430.
  • Azzam EI, De Toledo SM, Spitz DR, Little JB. 2002. Oxidative metabolism modulates signal transduction and micronucleus formation in bystander cells from alpha-particle-irradiated normal human fibroblast cultures. Cancer Res. 62:5436–5442.
  • Barcellos-Hoff MH, Brooks AL. 2001. Extracellular signaling through the microenvironment: a hypothesis relating carcinogenesis, bystander effects, and genomic instability. Radiat Res. 156:618–627
  • Barcellos-Hoff MH, Cucinotta FA. 2014. New tricks for an old fox: impact of TGFβ on the DNA damage response and genomic stability. Sci Signal. 7:re5.
  • Chai Y, Lam RK, Calaf GM, Zhou H, Amundson S, Hei TK. 2013. Radiation-induced non-targeted response in vivo: role of the TGFbeta-TGFBR1-COX-2 signalling pathway. Br J Cancer. 108:1106–1112.
  • Chaudhry MA, Omaruddin RA. 2012. Differential regulation of microRNA expression in irradiated and bystander cells. Mol Biol (Mosk). 46:634–643.
  • Denoyelle C, Lambert B, Meryet-Figuiere M, Vigneron N, Brotin E, Lecerf C, Abeilard E, Giffard F, Louis MH, Gauduchon P, et al. 2014. miR-491-5p-induced apoptosis in ovarian carcinoma depends on the direct inhibition of both BCL-XL and EGFR leading to BIM activation. Cell Death Dis. 5:e1445
  • Desai S, Srambikkal N, Yadav HD, Shetake N, Balla MM, Kumar A, Ray P, Ghosh A, Pandey BN. 2016. Molecular understanding of growth inhibitory effect from irradiated to bystander tumor cells in mouse fibrosarcoma tumor model. PLoS One. 11:e0161662.
  • Dinh TK, Fendler W, Chalubinska-Fendler J, Acharya SS, O'Leary C, Deraska PV, D'Andrea AD, Chowdhury D, Kozono D. 2016. Circulating miR-29a and miR-150 correlate with delivered dose during thoracic radiation therapy for non-small cell lung cancer. Radiat Oncol. 11:61.
  • Du S, Bouquet S, Lo CH, Pellicciotta I, Bolourchi S, Parry R, Barcellos-Hoff MH. 2015. Attenuation of the DNA damage response by transforming growth factor-beta inhibitors enhances radiation sensitivity of non-small-cell lung cancer cells in vitro and in vivo. Int J Radiat Oncol Biol Phys. 91:91–99.
  • Eldh M, Ekstrom K, Valadi H, Sjostrand M, Olsson B, Jernas M, Lotvall J. 2010. Exosomes communicate protective messages during oxidative stress; possible role of exosomal shuttle RNA. PLoS One. 5:e15353.
  • Georgakilas AG, Pavlopoulou A, Louka M, Nikitaki Z, Vorgias CE, Bagos PG, Michalopoulos I. 2015. Emerging molecular networks common in ionizing radiation, immune and inflammatory responses by employing bioinformatics approaches. Cancer Lett. 368:164–172.
  • Gilbert ES. 2009. Ionising radiation and cancer risks: what have we learned from epidemiology? Int J Radiat Biol. 85:467–482.
  • Gong F, Ren P, Zhang Y, Jiang J, Zhang H. 2016. MicroRNAs-491-5p suppresses cell proliferation and invasion by inhibiting IGF2BP1 in non-small cell lung cancer. Am J Transl Res. 8:485–495.
  • Guo R, Wang Y, Shi WY, Liu B, Hou SQ, Liu L. 2012. MicroRNA miR-491-5p targeting both TP53 and Bcl-XL induces cell apoptosis in SW1990 pancreatic cancer cells through mitochondria mediated pathway. Molecules. 17:14733–14747.
  • Haglund L, Bernier SM, Onnerfjord P, Recklies AD. 2008. Proteomic analysis of the LPS-induced stress response in rat chondrocytes reveals induction of innate immune response components in articular cartilage. Matrix Biol. 27:107–118.
  • Havaki S, Kotsinas A, Chronopoulos E, Kletsas D, Georgakilas A, Gorgoulis VG. 2015. The role of oxidative DNA damage in radiation induced bystander effect. Cancer Lett. 356:43–51.
  • Hu W, Xu S, Yao B, Hong M, Wu X, Pei H, Chang L, Ding N, Gao X, Ye C, et al. 2014. MiR-663 inhibits radiation-induced bystander effects by targeting TGFB1 in a feedback mode. RNA Biol. 11:1189–1198.
  • Hui Z, Yiling C, Wenting Y, XuQun H, ChuanYi Z, Hui L. 2015. miR-491-5p functions as a tumor suppressor by targeting JMJD2B in ERalpha-positive breast cancer. FEBS Lett. 589:812–821.
  • Isik Balci Y, Nuray E, Polat A, Enli Y, Ozgurler F, Akin M. 2016. Pentraxin-3 levels in beta thalassemia major and minor patients and its relationship with antioxidant capacity and total oxidant stress. J Pediat Hematol/Oncol. 38:12–16.
  • Jacob NK, Cooley JV, Yee TN, Jacob J, Alder H, Wickramasinghe P, Maclean KH, Chakravarti A. 2013. Identification of sensitive serum microRNA biomarkers for radiation biodosimetry. PLoS One. 8:e57603.
  • Jella KK, Rani S, O'Driscoll L, McClean B, Byrne HJ, Lyng FM. 2014. Exosomes are involved in mediating radiation induced bystander signaling in human keratinocyte cells. Radiat Res. 181:138–145.
  • Jelonek K, Widlak P, Pietrowska M. 2016. The influence of ionizing radiation on exosome composition, secretion and intercellular communication. Protein Peptide Lett. 23:656–663.
  • Jelonek K, Wojakowska A, Marczak L, Muer A, Tinhofer-Keilholz I, Lysek-Gladysinska M, Widlak P, Pietrowska M. 2015. Ionizing radiation affects protein composition of exosomes secreted in vitro from head and neck squamous cell carcinoma. Acta Biochimica Polonica. 62:265–272.
  • Jiang Y, Chen X, Tian W, Yin X, Wang J, Yang H. 2014. The role of TGF-beta1-miR-21-ROS pathway in bystander responses induced by irradiated non-small-cell lung cancer cells. Br J Cancer. 111:772–780.
  • Jin X, Chen Y, Chen H, Fei S, Chen D, Cai X, Liu L, Lin B, Su H, Zhao L, et al. 2017. Evaluation of tumor-derived exosomal miRNA as potential diagnostic biomarkers for early-stage non-small cell lung cancer using next-generation sequencing. Clin Cancer Res. 23:5311–5319.
  • Kadhim M, Salomaa S, Wright E, Hildebrandt G, Belyakov OV, Prise KM, Little MP. 2013. Non-targeted effects of ionising radiation-implications for low dose risk. Mutat Res. 752:84–98.
  • Kesminiene A, Evrard AS, Ivanov VK, Malakhova IV, Kurtinaitis J, Stengrevics A, Tekkel M, Anspaugh LR, Bouville A, Chekin S, et al. 2008. Risk of hematological malignancies among Chernobyl liquidators. Radiat Res. 170:721–735.
  • Kulkarni S, Koller A, Mani KM, Wen R, Alfieri A, Saha S, Wang J, Patel P, Bandeira N, Guha C, et al. 2016. Identifying urinary and serum exosome biomarkers for radiation exposure using a data dependent acquisition and SWATH-MS combined workflow. Int J Radiat Oncol Biol Phys. 96:566–577.
  • Laurent O, Ancelet S, Richardson DB, Hemon D, Ielsch G, Demoury C, Clavel J, Laurier D. 2013. Potential impacts of radon, terrestrial gamma and cosmic rays on childhood leukemia in France: a quantitative risk assessment. Radiat Environ Biophys. 52:195–209.
  • Laurier D, Richardson DB, Cardis E, Daniels RD, Gillies M, O'Hagan J, Hamra GB, Haylock R, Leuraud K, Moissonnier M, et al. 2017. The international nuclear workers study (Inworks): a collaborative epidemiological study to improve knowledge about health effects of protracted low-dose exposure. Radiat Prot Dosimetry. 173:21–25.
  • Liu SZ, Jin SZ, Liu XD. 2004. Radiation-induced bystander effect in immune response. Biomed Environ Sci. 17:40–46.
  • Lorimore SA, Chrystal JA, Robinson JI, Coates PJ, Wright EG. 2008. Chromosomal instability in unirradiated hemaopoietic cells induced by macrophages exposed in vivo to ionizing radiation. Cancer Res. 68:8122–8126.
  • Lotvall J, Hill AF, Hochberg F, Buzas EI, Di Vizio D, Gardiner C, Gho YS, Kurochkin IV, Mathivanan S, Quesenberry P, 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
  • Lyng FM, Maguire P, McClean B, Seymour C, Mothersill C. 2006. The involvement of calcium and MAP kinase signaling pathways in the production of radiation-induced bystander effects. Radiat Res. 165:400–409.
  • Lyng FM, Seymour CB, Mothersill C. 2000. Production of a signal by irradiated cells which leads to a response in unirradiated cells characteristic of initiation of apoptosis. Br J Cancer. 83:1223–1230.
  • Malla B, Zaugg K, Vassella E, Aebersold DM, Dal Pra A. 2017. Exosomes and exosomal microRNAs in prostate cancer radiation therapy. Int J Radiat Oncol Biol Phys. 98:982–995.
  • Mantovani A, Garlanda C, Bottazzi B. 2003. Pentraxin 3, a non-redundant soluble pattern recognition receptor involved in innate immunity. Vaccine. 21(Suppl 2):S43–S47.
  • Matsumura S, Demaria S. 2010. Up-regulation of the pro-inflammatory chemokine CXCL16 is a common response of tumor cells to ionizing radiation. Radiat Res. 173:418–425.
  • Menon N, Rogers CJ, Lukaszewicz AI, Axtelle J, Yadav M, Song F, Chakravarti A, Jacob NK. 2016. Detection of acute radiation sickness: a feasibility study in non-human primates circulating miRNAs for triage in radiological events. PLoS One. 11:e0167333.
  • Mole RH, Papworth DG, Corp MJ. 1983. The dose-response for x-ray induction of myeloid leukaemia in male CBA/H mice. Br J Cancer. 47:285–291.
  • Mutschelknaus L, Peters C, Winkler K, Yentrapalli R, Heider T, Atkinson MJ, Moertl S. 2016. Exosomes Derived from squamous head and neck cancer promote cell survival after ionizing radiation. PLoS One. 11:e0152213.
  • Nagasawa H, Huo L, Little JB. 2003. Increased bystander mutagenic effect in DNA double-strand break repair-deficient mammalian cells. Int J Radiat Biol. 79:35–41.
  • Najafi M, Fardid R, Hadadi G, Fardid M. 2014. The mechanisms of radiation-induced bystander effect. J Biomed Phys Eng. 4:163–172.
  • Nikitaki Z, Mavragani IV, Laskaratou DA, Gika V, Moskvin VP, Theofilatos K, Vougas K, Stewart RD, Georgakilas AG. 2016. Systemic mechanisms and effects of ionizing radiation: a new ‘old’ paradigm of how the bystanders and distant can become the players. Sem Cancer Biol. 37–38:77–95.
  • Pearce MS, Salotti JA, Little MP, McHugh K, Lee C, Kim KP, Howe NL, Ronckers CM, Rajaraman P, Sir Craft AW, et al. 2012. Radiation exposure from CT scans in childhood and subsequent risk of leukaemia and brain tumours: a retrospective cohort study. Lancet (London, England). 380: 9840–9499. 505.
  • Pixley FJ, Stanley ER. 2004. CSF-1 regulation of the wandering macrophage: complexity in action. Trends Cell Biol. 14:628–638.
  • Prise KM, Folkard M, Michael BD. 2003. A review of the bystander effect and its implications for low-dose exposure. Radiat Prot Dosimetry. 104:347–355.
  • Rodel F, Frey B, Gaipl U, Keilholz L, Fournier C, Manda K, Schollnberger H, Hildebrandt G, Rodel C. 2012. Modulation of inflammatory immune reactions by low-dose ionizing radiation: molecular mechanisms and clinical application. Curr Med Chem. 19:1741–1750.
  • Rodel F, Frey B, Multhoff G, Gaipl U. 2015. Contribution of the immune system to bystander and non-targeted effects of ionizing radiation. Cancer Lett. 356:105–113.
  • Schroeder T, Geyh S, Germing U, Haas R. 2016. Mesenchymal stromal cells in myeloid malignancies. Blood Res. 51:225–232.
  • Shao C, Folkard M, Prise KM. 2008. Role of TGF-beta1 and nitric oxide in the bystander response of irradiated glioma cells. Oncogene. 27:434–440.
  • Shiraki A, Oyama J, Komoda H, Asaka M, Komatsu A, Sakuma M, Kodama K, Sakamoto Y, Kotooka N, Hirase T, et al. 2012. The glucagon-like peptide 1 analog liraglutide reduces TNF-alpha-induced oxidative stress and inflammation in endothelial cells. Atherosclerosis. 221:375–382.
  • Sproull M, Kramp T, Tandle A, Shankavaram U, Camphausen K. 2017. Multivariate analysis of radiation responsive proteins to predict radiation exposure in total-body irradiation and partial-body irradiation models. Radiat Res. 187:251–258.
  • Sugihara T, Murano H, Nakamura M, Tanaka K. 2013. In vivo partial bystander study in a mouse model by chronic medium-dose-rate gamma-ray irradiation. Radiat Res. 179:221–231.
  • Sun R, Liu Z, Tong D, Yang Y, Guo B, Wang X, Zhao L, Huang C. 2017. miR-491-5p, mediated by Foxi1, functions as a tumor suppressor by targeting Wnt3a/beta-catenin signaling in the development of gastric cancer. Cell Death Dis. 8:e2714.
  • Szatmari T, Kis D, Bogdandi EN, Benedek A, Bright S, Bowler D, Persa E, Kis E, Balogh A, Naszalyi LN, et al. 2017. Extracellular vesicles mediate radiation-induced systemic bystander signals in the bone marrow and spleen. Front Immunol. 8:347
  • Tada N, Tsuno NH, Kawai K, Murono K, Nirei T, Ishihara S, Sunami E, Kitayama J, Watanabe T. 2014. Changes in the plasma levels of cytokines/chemokines for predicting the response to chemoradiation therapy in rectal cancer patients. Oncology Rep. 31:463–471.
  • Tomandlova M, Jarkovsky J, Tomandl J, Kubkova L, Kala P, Littnerova S, Gottwaldova J, Kubena P, Ganovska E, Poloczek M, et al. 2015. Prognostic value of pentraxin-3 level in patients with STEMI and its relationship with heart failure and markers of oxidative stress. Dis Markers. 2015:159051.
  • van der Pol E, Boing AN, Harrison P, Sturk A, Nieuwland R. 2012. Classification, functions, and clinical relevance of extracellular vesicles. Pharmacol Rev. 64:676–705.
  • Vlachos IS, Paraskevopoulou MD, Karagkouni D, Georgakilas G, Vergoulis T, Kanellos I, Anastasopoulos IL, Maniou S, Karathanou K, Kalfakakou D, et al. 2015. DIANA-TarBase v7.0: indexing more than half a million experimentally supported miRNA:mRNA interactions. Nucleic Acids Res. 43:D153–D159.
  • Vlachos IS, Zagganas K, Paraskevopoulou MD, Georgakilas G, Karagkouni D, Vergoulis T, Dalamagas T, Hatzigeorgiou AG. 2015. DIANA-miRPath v3.0: deciphering microRNA function with experimental support. Nucleic Acids Res. 43:W460–W466.
  • Widlak P, Jelonek K, Wojakowska A, Pietrowska M, Polanska J, Marczak L, Miszczyk L, Skladowski K. 2015. Serum proteome signature of radiation response: upregulation of inflammation-related factors and downregulation of apolipoproteins and coagulation factors in cancer patients treated with radiation therapy – a pilot study. Int J Radiat Oncol Biol Phys. 92:1108–1115.
  • Xu S, Ding N, Pei H, Hu W, Wei W, Zhang X, Zhou G, Wang J. 2014. MiR-21 is involved in radiation-induced bystander effects. RNA Biol. 11:1161–1170.
  • Xu S, Wang J, Ding N, Hu W, Zhang X, Wang B, Hua J, Wei W, Zhu Q. 2015. Exosome-mediated microRNA transfer plays a role in radiation-induced bystander effect. RNA Biol. 12:1355–1363.
  • Yentrapalli R, Merl-Pham J, Azimzadeh O, Mutschelknaus L, Peters C, Hauck SM, Atkinson MJ, Tapio S, Moertl S. 2017. Quantitative changes in the protein and miRNA cargo of plasma exosome-like vesicles after exposure to ionizing radiation. Int J Radiat Biol. 93:569–580.
  • Yin X, Tian W, Wang L, Wang J, Zhang S, Cao J, Yang H. 2015. Radiation quality-dependence of bystander effect in unirradiated fibroblasts is associated with TGF-beta1-Smad2 pathway and miR-21 in irradiated keratinocytes. Sci Rep. 5:11373.
  • Yoon MS, Pham CT, Phan MT, Shin DJ, Jang YY, Park MH, Kim SK, Kim S, Cho D. 2016. Irradiation of breast cancer cells enhances CXCL16 ligand expression and induces the migration of natural killer cells expressing the CXCR6 receptor. Cytotherapy. 18:1532–1542.
  • Yu T, Zuo QF, Gong L, Wang LN, Zou QM, Xiao B. 2016. MicroRNA-491 regulates the proliferation and apoptosis of CD8(+) T cells. Sci Rep. 6:30923
  • Zhou Y, Li Y, Ye J, Jiang R, Yan H, Yang X, Liu Q, Zhang J. 2013. MicroRNA-491 is involved in metastasis of hepatocellular carcinoma by inhibitions of matrix metalloproteinase and epithelial to mesenchymal transition. Liver Int. 33:1271–1280.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.