Platelet RNA signatures for the detection of cancer

Nik Sol, Thomas Wurdinger, Nik Sol, Thomas Wurdinger

Abstract

Platelets are equipped with RNA processing machineries, such as pre-mRNA splicing, pre-miRNA processing, and mRNA translation. Since platelets are devoid of a nucleus, most RNA transcripts in platelets are derived from megakaryocytes during thrombocytogenesis. However, platelets can also ingest RNA molecules during circulation and/or interaction with other cell types. Since platelets were first described by Bizzozero in 1881, their well-established role in hemostasis and thrombosis has been intensively studied. However, in the past decades, the list of biological processes in which platelets play an important role keeps expanding. In this review, we discuss how platelet RNA biomarker signatures can be altered in the presence of cancer.

Keywords: Biomarkers; Liquid biopsy; Platelets; Splicing; Transcriptome; Tumor-educated platelets; mRNA.

Figures

Fig. 1
Fig. 1
Schematic representation of tumor-mediated education of platelets and the megakaryocyte leading to metastasis. (a) Megakaryocytes in the bone marrow and lungs sort specific RNA and proteins into platelet precursors. (b) Circulating platelets contain a variety of RNA transcripts and proteins. During their 7–10-day lifespan, platelets interact with immune cells, cancer cells, and stromal cells. These direct interactions as well as distant cell signaling, for instance, via vesicle-mediated communication in whole blood, changes the content of the platelet and platelet function. (c) This process leads to the development of tumor-educated platelets. Next, tumor-educated platelets can influence the process of metastasis formation by stimulating or blocking immune cells, endothelial cells, stromal cells, and cancer cells, either by direct cell-to-cell contact or by releasing extracellular queues. (d) Finally, metastasis could affect the sorting of specific RNA and proteins of megakaryocytes into platelets
Fig. 2
Fig. 2
ThromboSeq workflow. (a) TEP mRNA sequencing workflow; blood draw is performed on patients, and from a single 6-ml EDTA-coated tube, platelets are isolated. RNA extraction is done according to manufacturer’s protocol using the mirVana RNA isolation kit (Life Technologies). Next, mRNA is amplified using SMARTer Ultra low input RNA kit (Clontech). Samples are prepared for sequencing on the Hiseq 2500 Illumina platform using the Truseq Nano DNA Sample Prep Kit (Illumina). After each step, quality control was performed by critical inspection of Bioanalyzer profiles (Agilent). (b) Heatmap of unsupervised clustering of platelet mRNA profiles of healthy donors (red, n = 55) and patients with cancer (gray, n = 228)
Fig. 3
Fig. 3
Correlation plots of TEP RNA signatures with other (a) nucleated blood cells. Correlation plots between platelets sequenced by Best et al. compared with platelet RNA expression levels from different studies and compared with RNA expression from different blood cells. (a) Platelets from Bray et al. [82]. (b) Poly A selected RNAs from platelets from Kissopoulou et al. [83]. (c) Ribosomal RNA-depleted platelets from Kissopoulou et al. [83]. (d) Platelets from Rowley et al. [84]. (e) Platelets from Simon et al. [85]. (f) Megakaryocyte [86]. (g) Memory T-cells [87]. (h) CD4 T-cells [87]. (i) CD8 T-cells [87]. (j) Granulocytes [87]. (k) B-cells [87]. (l) Monocytes [87]. (m) Natural killer cells [87]

References

    1. McAllister SS, Weinberg RA. The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis. Nature Cell Biology. 2014;16:717–727. doi: 10.1038/ncb3015.
    1. Kuznetsov HS, et al. Identification of luminal breast cancers that establish a tumor-supportive macroenvironment defined by proangiogenic platelets and bone marrow-derived cells. Cancer Discovery. 2012;2:1150–1165. doi: 10.1158/-12-0216.
    1. Nilsson RJA, et al. Blood platelets contain tumor-derived RNA biomarkers. Blood. 2011;118:3680–3683. doi: 10.1182/blood-2011-03-344408.
    1. Best MG, et al. RNA-Seq of tumor-educated platelets enables blood-based pan-cancer, multiclass, and molecular pathway cancer diagnostics. Cancer Cell. 2015;28:666–676. doi: 10.1016/j.ccell.2015.09.018.
    1. Calverley DC, et al. Significant downregulation of platelet gene expression in metastatic lung cancer. Clinical and Translational Science. 2010;3:227–232. doi: 10.1111/j.1752-8062.2010.00226.x.
    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013.
    1. Borsig L. The role of platelet activation in tumor metastasis. Expert Review of Anticancer Therapy. 2008;8:1247–1255. doi: 10.1586/14737140.8.8.1247.
    1. Steinert BW, Tang DG, Grossi IM, Umbarger LA, Honn KV. Studies on the role of platelet eicosanoid metabolism and integrin alpha IIb beta 3 in tumor-cell-induced platelet aggregation. International Journal of Cancer. 1993;54:92–101. doi: 10.1002/ijc.2910540116.
    1. Zucchella, M., et al. (1989). Human tumor cells cultured “in vitro” activate platelet function by producing ADP or thrombin. Haematologica, 74, 541–545.
    1. Egan K, et al. Platelet adhesion and degranulation induce pro-survival and pro-angiogenic signalling in ovarian cancer cells. PloS One. 2011;6:e26125. doi: 10.1371/journal.pone.0026125.
    1. Menter DG, et al. Platelets and cancer: a casual or causal relationship: revisited. Cancer Metastasis Reviews. 2014;33:231–269. doi: 10.1007/s10555-014-9498-0.
    1. Sierko E, Wojtukiewicz MZ. Platelets and angiogenesis in malignancy. Seminars in Thrombosis and Hemostasis. 2004;30:95–108. doi: 10.1055/s-2004-822974.
    1. Placke T, et al. Platelet-derived MHC class I confers a pseudonormal phenotype to cancer cells that subverts the antitumor reactivity of natural killer immune cells. Cancer Research. 2012;72:440–448. doi: 10.1158/0008-5472.CAN-11-1872.
    1. Borsig L, et al. Heparin and cancer revisited: mechanistic connections involving platelets, P-selectin, carcinoma mucins, and tumor metastasis. Proceedings of the National Academy of Sciences of the United States of America. 2001;98:3352–3357. doi: 10.1073/pnas.061615598.
    1. Palumbo JS, et al. Platelets and fibrin(ogen) increase metastatic potential by impeding natural killer cell-mediated elimination of tumor cells. Blood. 2005;105:178–185. doi: 10.1182/blood-2004-06-2272.
    1. Palumbo JS, et al. Tumor cell-associated tissue factor and circulating hemostatic factors cooperate to increase metastatic potential through natural killer cell-dependent and-independent mechanisms. Blood. 2007;110:133–141. doi: 10.1182/blood-2007-01-065995.
    1. Labelle M, Begum S, Hynes RO. Direct signaling between platelets and cancer cells induces an epithelial-mesenchymal-like transition and promotes metastasis. Cancer Cell. 2011;20:576–590. doi: 10.1016/j.ccr.2011.09.009.
    1. Buergy D, Wenz F, Groden C, Brockmann MA. Tumor-platelet interaction in solid tumors. International Journal of Cancer. 2012;130:2747–2760. doi: 10.1002/ijc.27441.
    1. Gasic GJ, Gasic TB, Stewart CC. Antimetastatic effects associated with platelet reduction. Proceedings of the National Academy of Sciences of the United States of America. 1968;61:46–52. doi: 10.1073/pnas.61.1.46.
    1. Alix-Panabières C, Pantel K. Clinical applications of circulating tumor cells and circulating tumor DNA as liquid biopsy. Cancer Discovery. 2016;6:479–491. doi: 10.1158/-15-1483.
    1. Diaz LA, Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. Journal of Clinical Oncology. 2014;32:579–586. doi: 10.1200/JCO.2012.45.2011.
    1. Bardelli A, Pantel K. Liquid biopsies, what we do not know (yet) Cancer Cell. 2017;31:172–179. doi: 10.1016/j.ccell.2017.01.002.
    1. Chu, D., et al. (2016). ESR1 mutations in circulating plasma tumor DNA from metastatic breast cancer patients. Clinical Cancer Research, 22(4), 993–999.
    1. Guttery, D. S., et al. (2015). Noninvasive detection of activating estrogen receptor 1 (ESR1) mutations in estrogen receptor–positive metastatic breast cancer. Clinical Chemistry, 61(7), 974–82.
    1. Schiavon, G., et al. (2015). Analysis of ESR1 mutation in circulating tumor DNA demonstrates evolution during therapy for metastatic breast cancer. Science Translational Medicine, 7(313), 313ra182.
    1. Gorges TM, et al. Heterogeneous PSMA expression on circulating tumor cells: a potential basis for stratification and monitoring of PSMA-directed therapies in prostate cancer. Oncotarget. 2016;7:34930–34941. doi: 10.18632/oncotarget.9004.
    1. Gorges, T. M., et al. (2016). Enumeration and molecular characterization of tumor cells in lung cancer patients using a novel in vivo device for capturing circulating tumor cells. Clinical Cancer Research, 22(9), 2197–206.
    1. Maheswaran S, et al. Detection of mutations in EGFR in circulating lung-cancer cells. The New England Journal of Medicine. 2008;359:366–377. doi: 10.1056/NEJMoa0800668.
    1. Miyamoto, D. T., et al. (2012). Androgen receptor signaling in circulating tumor cells as a marker of hormonally responsive prostate cancer. Cancer Discovery, 2(11), 995–1003.
    1. Antonarakis ES, et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. The New England Journal of Medicine. 2014;371:1028–1038. doi: 10.1056/NEJMoa1315815.
    1. Siravegna G, et al. Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nature Medicine. 2015;21:795–801. doi: 10.1038/nm.3870.
    1. Gerlinger M, Norton L, Swanton C. Acquired resistance to crizotinib from a mutation in CD74-ROS1. The New England Journal of Medicine. 2013;369:1172–1173. doi: 10.1056/NEJMc1309091.
    1. Nilsson RJA, et al. Rearranged EML4-ALK fusion transcripts sequester in circulating blood platelets and enable blood-based crizotinib response monitoring in non-small-cell lung cancer. Oncotarget. 2015;7:1066–1075. doi: 10.18632/oncotarget.6279.
    1. Schwarzenbach H, Pantel K. Circulating DNA as biomarker in breast cancer. Breast Cancer Research. 2015;17:136. doi: 10.1186/s13058-015-0645-5.
    1. Alix-Panabières C, Pantel K. Circulating tumor cells: liquid biopsy of cancer. Clinical Chemistry. 2013;59:110–118. doi: 10.1373/clinchem.2012.194258.
    1. Alix-Panabières C, Pantel K. Challenges in circulating tumour cell research. Nature Reviews. Cancer. 2014;14:623–631. doi: 10.1038/nrc3820.
    1. Melo SA, et al. Glypican−1 identifies cancer exosomes and detects early pancreatic cancer. Nature. 2015;523:177–182. doi: 10.1038/nature14581.
    1. Skog J, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nature Cell Biology. 2008;10:1470–1476. doi: 10.1038/ncb1800.
    1. Stone RL, et al. Paraneoplastic thrombocytosis in ovarian cancer. The New England Journal of Medicine. 2012;366:610–618. doi: 10.1056/NEJMoa1110352.
    1. Mantur M, Koper O, Snarska J, Sidorska A, Kruszewska-Wnorowska K. Evaluation of {PDGF}-{AB} and {sP}-selectin concentrations in relation to platelet count in patients with colorectal cancer before and after surgical treatment. Polskie Archiwum Medycyny Wewnętrznej. 2008;118:345–350.
    1. Cho MS, et al. Platelets increase the proliferation of ovarian cancer cells. Blood. 2012;120:4869–4872. doi: 10.1182/blood-2012-06-438598.
    1. Bottsford-Miller J, et al. Differential platelet levels affect response to taxane-based therapy in ovarian cancer. Clinical Cancer Research. 2015;21:602–610. doi: 10.1158/1078-0432.CCR-14-0870.
    1. Ryningen A, Apelseth T, Hausken T, Bruserud Ø. Reticulated platelets are increased in chronic myeloproliferative disorders, pure erythrocytosis, reactive thrombocytosis and prior to hematopoietic reconstitution after intensive chemotherapy. Platelets. 2006;17:296–302. doi: 10.1080/09537100600746508.
    1. Matowicka-Karna J, Kamocki Z, Polińska B, Osada J, Kemona H. Platelets and inflammatory markers in patients with gastric cancer. Clinical & Developmental Immunology. 2013;2013:1–6. doi: 10.1155/2013/401623.
    1. Wang R, et al. Electron cryotomography reveals ultrastructure alterations in platelets from patients with ovarian cancer. Proceedings of the National Academy of Sciences. 2015;112:14266–14271. doi: 10.1073/pnas.1518628112.
    1. Mantur M, Kemona H, Pietruczuk M, Wasiluk A. Does renal carcinoma affect the expression of P-selectin on platelets? Neoplasma. 2002;49:243–245.
    1. Mantur M, Kemona H, Kozłowski R, Kemona-Chetnik I. Effect of tumor stage and nephrectomy on CD62P expression and sP-selectin concentration in renal cancer. Neoplasma. 2003;50:262–265.
    1. Dymicka-Piekarska V, Matowicka-Karna J, Osada J, Kemona H, Butkiewicz AM. Changes in platelet CD 62P expression and soluble P-selectin concentration in surgically treated colorectal carcinoma. Advances in Medical Sciences. 2006;51:304–308.
    1. Osada J, Rusak M, Kamocki Z, Dabrowska MI, Kedra B. Platelet activation in patients with advanced gastric cancer. Neoplasma. 2010;57:145–150. doi: 10.4149/neo_2010_02_145.
    1. Peterson JE, et al. VEGF, PF4 and PDGF are elevated in platelets of colorectal cancer patients. Angiogenesis. 2012;15:265–273. doi: 10.1007/s10456-012-9259-z.
    1. Kamińska J, et al. Does thrombopoiesis in multiple myeloma patients depend on the stage of the disease? Advances in Medical Sciences. 2014;59:166–171. doi: 10.1016/j.advms.2013.12.006.
    1. Meikle CKS, et al. Cancer and thrombosis: the platelet perspective. Frontiers in Cell and Development Biology. 2017;4:147. doi: 10.3389/fcell.2016.00147.
    1. Cooke NM, et al. Increased platelet reactivity in patients with late-stage metastatic cancer. Cancer Medicine. 2013;2:564–570. doi: 10.1002/cam4.86.
    1. Angénieux C, et al. Time-dependent decay of mRNA and ribosomal RNA during platelet aging and its correlation with translation activity. PloS One. 2016;11:e0148064. doi: 10.1371/journal.pone.0148064.
    1. Balduini CL, et al. Relationship between size and thiazole orange fluorescence of platelets in patients undergoing high-dose chemotherapy. British Journal of Haematology. 1999;106:202–207. doi: 10.1046/j.1365-2141.1999.01475.x.
    1. Nilsson RJA, et al. Rearranged EML4-ALK fusion transcripts sequester in circulating blood platelets and enable blood-based crizotinib response monitoring in non-small-cell lung cancer. Oncotarget. 2010;1:1066–1075.
    1. Brown GT, McIntyre TM. Lipopolysaccharide signaling without a nucleus: kinase cascades stimulate platelet shedding of proinflammatory IL-1β-rich microparticles. Journal of Immunology. 2011;186:5489–5496. doi: 10.4049/jimmunol.1001623.
    1. Chen, S., et al. (2016). Biological function and mechanism of MALAT-1 in renal cell carcinoma proliferation and apoptosis: role of the MALAT-1–Livin protein interaction. The Journal of Physiological Sciences. doi:10.1007/s12576-016-0486-8.
    1. Huang J, et al. MALAT1 promotes the proliferation and invasion of thyroid cancer cells via regulating the expression of IQGAP1. Biomedicine & Pharmacotherapy. 2016;83:1–7. doi: 10.1016/j.biopha.2016.05.039.
    1. Chen D, et al. The role of MALAT-1 in the invasion and metastasis of gastric cancer. Scandinavian Journal of Gastroenterology. 2017;52:790–796. doi: 10.1080/00365521.2017.1280531.
    1. Bian D, Shi W, Shao Y, Li P, Song G. Long non-coding RNA GAS5 inhibits tumorigenesis via miR-137 in melanoma. American Journal of Translational Research. 2017;9:1509–1520.
    1. Luo, G., et al. (2017). LncRNA GAS5 inhibits cellular proliferation by targeting P27Kip1. Molecular Cancer Research. doi:10.1158/1541-7786 MCR-16-0331.
    1. Huang T, et al. SNHG8 is identified as a key regulator of epstein-barr virus(EBV)-associated gastric cancer by an integrative analysis of lncRNA and mRNA expression. Oncotarget. 2016;7:80990–81002.
    1. Zhao L, et al. Long non-coding RNA SNHG5 suppresses gastric cancer progression by trapping MTA2 in the cytosol. Oncogene. 2016;35:5770–5780. doi: 10.1038/onc.2016.110.
    1. Zhao L, et al. The lncRNA SNHG5/miR-32 axis regulates gastric cancer cell proliferation and migration by targeting KLF4. The FASEB Journal. 2017;31:893–903. doi: 10.1096/fj.201600994R.
    1. Russell MR, et al. CASC15-S is a tumor suppressor lncRNA at the 6p22 neuroblastoma susceptibility locus. Cancer Research. 2015;75:3155–3166. doi: 10.1158/0008-5472.CAN-14-3613.
    1. Morenos L, et al. Hypermethylation and down-regulation of DLEU2 in paediatric acute myeloid leukaemia independent of embedded tumour suppressor miR-15a/16-1. Molecular Cancer. 2014;13:123. doi: 10.1186/1476-4598-13-123.
    1. Kasar S, et al. Therapeutic implications of activation of the host gene (Dleu2) promoter for miR-15a/16-1 in chronic lymphocytic leukemia. Oncogene. 2014;33:3307–3315. doi: 10.1038/onc.2013.291.
    1. Plé H, et al. The repertoire and features of human platelet microRNAs. PloS One. 2012;7:e50746. doi: 10.1371/journal.pone.0050746.
    1. Dangwal S, Thum T. MicroRNAs in platelet physiology and pathology. Hämostaseologie. 2013;33:17–20. doi: 10.5482/HAMO-13-01-0002.
    1. Dangwal S, Thum T. MicroRNAs in platelet biogenesis and function. Thrombosis and Haemostasis. 2012;108:599–604. doi: 10.1160/TH12-03-0211.
    1. Edelstein LC, Bray PF. MicroRNAs in platelet production and activation. Blood. 2011;117:5289–5296. doi: 10.1182/blood-2011-01-292011.
    1. Edelstein LC, et al. MicroRNAs in platelet production and activation. Journal of Thrombosis and Haemostasis. 2013;11(Suppl 1):340–350. doi: 10.1111/jth.12214.
    1. Gidlöf O, et al. Platelets activated during myocardial infarction release functional miRNA, which can be taken up by endothelial cells and regulate ICAM1 expression. Blood. 2013;121:3908–3917. doi: 10.1182/blood-2012-10-461798.
    1. Gnatenko DV, et al. Class prediction models of thrombocytosis using genetic biomarkers. Blood. 2009;115:7–14. doi: 10.1182/blood-2009-05-224477.
    1. Healy AM, et al. Platelet expression profiling and clinical validation of myeloid-related protein-14 as a novel determinant of cardiovascular events. Circulation. 2006;113:2278–2284. doi: 10.1161/CIRCULATIONAHA.105.607333.
    1. Lood C, et al. Platelet transcriptional profile and protein expression in patients with systemic lupus erythematosus: up-regulation of the type I interferon system is strongly associated with vascular disease. Blood. 2010;116:1951–1957. doi: 10.1182/blood-2010-03-274605.
    1. Raghavachari N, et al. Amplified expression profiling of platelet transcriptome reveals changes in arginine metabolic pathways in patients with sickle cell disease. Circulation. 2007;115:1551–1562. doi: 10.1161/CIRCULATIONAHA.106.658641.
    1. Rauschenberger L, et al. Exosomal particles secreted by prostate cancer cells are potent mRNA and protein vehicles for the interference of tumor and tumor environment. Prostate. 2016;76:409–424. doi: 10.1002/pros.23132.
    1. Moscardó A, Latorre A, Santos MT, Bonanad S, Vallés J. Platelet function in malignant hematological disorders. Current Opinion in Oncology. 2015;27:522–531. doi: 10.1097/CCO.0000000000000237.
    1. Pantel K, Alix-Panabieres C. Functional studies on viable circulating tumor cells. Clinical Chemistry. 2016;62:328–334. doi: 10.1373/clinchem.2015.242537.
    1. Bray PF, et al. The complex transcriptional landscape of the anucleate human platelet. BMC Genomics. 2013;14:1. doi: 10.1186/1471-2164-14-1.
    1. Kissopoulou A, Jonasson J, Lindahl TL, Osman A. Next generation sequencing analysis of human platelet PolyA+ mRNAs and rRNA-depleted total RNA. PloS One. 2013;8:e81809. doi: 10.1371/journal.pone.0081809.
    1. Rowley JW, et al. Genome-wide RNA-seq analysis of human and mouse platelet transcriptomes. Blood. 2011;118:e101–e111. doi: 10.1182/blood-2011-03-339705.
    1. Simon LM, et al. Human platelet microRNA-mRNA networks associated with age and gender revealed by integrated plateletomics. Blood. 2014;123:e37–e45. doi: 10.1182/blood-2013-12-544692.
    1. Chen L, et al. Transcriptional diversity during lineage commitment of human blood progenitors. Science. 2014;345:1251033. doi: 10.1126/science.1251033.
    1. Hrdlickova B, et al. Expression profiles of long non-coding RNAs located in autoimmune disease-associated regions reveal immune cell-type specificity. Genome Medicine. 2014;6:88. doi: 10.1186/s13073-014-0088-0.

Source: PubMed

3
Abonneren