Exosomes from Plasmodium-infected hosts inhibit tumor angiogenesis in a murine Lewis lung cancer model

Y Yang, Q Liu, J Lu, D Adah, S Yu, S Zhao, Y Yao, L Qin, L Qin, X Chen, Y Yang, Q Liu, J Lu, D Adah, S Yu, S Zhao, Y Yao, L Qin, L Qin, X Chen

Abstract

Previous research to investigate the interaction between malaria infection and tumor progression has revealed that malaria infection can potentiate host immune response against tumor in tumor-bearing mice. Exosomes may play key roles in disseminating pathogenic host-derived molecules during infection because several studies have shown the involvement and roles of extracellular vesicles in cell-cell communication. However, the role of exosomes generated during Plasmodium infection in tumor growth, progression and angiogenesis has not been studied either in animals or in the clinics. To test this hypothesis, we designed an animal model to generate and isolate exosomes from mice which were subsequently used to treat the tumor. Intra-tumor injection of exosomes derived from the plasma of Plasmodium-infected mice provided significantly reduced Lewis lung cancer growth in mice. We further co-cultured the isolated exosomes with endothelial cells and observed significantly reduced expression of VEGFR2 and migration in the endothelial cells. Interestingly, high level of micro-RNA (miRNA) 16/322/497/17 was detected in the exosomes derived from the plasma of mice infected with Plasmodium compared with those from control mice. We observed that overexpression of the miRNA 16/322/497/17 in endothelial cell corresponded with decreased expression of VEGFR2, inhibition of angiogenesis and inhibition of the miRNA 16/322/497/17 significantly alleviated these effects. These data provide novel scientific evidence of the interaction between Plasmodium infection and lung cancer growth and angiogenesis.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Identification of exosomes characteristics. (a) Using Nanosight to detect particle numbers of exosomes isolated from plasma of mice in different groups (*P<0.05, compared with naïve ex and LLC ex groups). (b) Using Nanosight to detect size of exosome isolated from plasma of mice in different groups. (c) A representative electron microscopic image of exosomes from mice plasma; scale bar 200 nm. (d) Confirmed by western blot for CD63, CD9, CD81 and Hsp70 exosomes markers from plasma of mice.
Figure 2
Figure 2
Exosomes suppressed tumor growth. (a) Longitudinal exosomes intra-tumor injection (each groups n=6). (b) C57BL/6 mice were inoculated with 5 × 105 lewis lung cells subcutaneously. Exosomes were injected intra-tumor from day 7 when tumor reached 3 × 3 mm2 in size. Tumor growth was monitored (V=ab2/2). Bars correspond to mean±s.d. (*P<0.05 Naïve ex versus Py ex; LLC ex versus Py+LLC ex).
Figure 3
Figure 3
Exosomes inhibited angiogenesis. (a) At 19 days, tumors were collected and presented as shown. (b) Tumors displayed varying degrees of feeding tumor vessels along the undersurface of the surrounding dermis at 19 days post inoculation. Quantification of blood vessels feeding to tumors treated with exosomes of different groups, or PBS. n=5. (*P<0.05). (c) Immunohistochemical analysis of CD31 expression in tumor tissues. Scale bar 20 μm (***P<0.001 Py ex and Py+LLC ex groups compared with PBS, Naïve ex and LLC ex groups).
Figure 4
Figure 4
Exosomes uptake by MS1 cells in vitro. (a) Confocal images of cultures exposed to exosomes at 6 h. The red channel is representative of Dir emission, the blue channel is Hoechst nuclear stain. Fluorescent signals are merged with transmission images. Scale bar is 20 μm. (b) MS1 co-culture with different exosomes at 24 h and uptake confirmed by western blot for red blood cell special marker CD235a.
Figure 5
Figure 5
The effect of exosomes on tube formation and migration in endothelial cell. (a) Endothelial cell tube formation assay showed interference of network assembly of MS1 cells on pre-solidified Matrigel in medium containing exosomes. Scale bar 500 μm (*P<0.05). (b) The invasion activity of MS1 cell incubated with different exosomes using transwell method. Scale bar 100 μm (NC: Negative control, PC: Positive control, ***P<0.001). (c) Endothelial cell migration was measured after co-culture with different groups exosomes for 24 h. Lines indicated the edge of the ‘wound’ directly after making the scratch. Statistical analysis of the width of the scratches is shown on the right (*P<0.05).
Figure 6
Figure 6
Exosomes inhibited vegfR2 expression in MS1 cells. (a) MS1 cells were cultured in vitro and exosomes were added in the culture medium for 24 h. VEGFR2 mRNA expression was detected using qPCR (*P<0.05). (b) Western blotting analysis of phospho-VEGFR2 and VEGFR2 expression in MS1 cells co-culture with exosomes; Quantitative results of western blotting assay showed thatVEGFR2 and phospho-VEGFR2 expression (*P<0.05).
Figure 7
Figure 7
miRNAs is overexpressed in plasmodium-infected mice plasma exosomes, downregulated VEGFR-2 and inhibited tube formation. (a) qPCR detected the level of miRNAs expression in exosomes of four groups. (b) VEGFR2 is a target gene of miR(16-5p/322-5p/497-5p/17-5p). (c) Luciferase reporter assay was performed using 293T cells as described in the Materials and methods section (*P<0.05,compared with miRNA negative control; # P<0.05, compared with miRNAs add inhibitors). (d) The relative expression of VEGFR2 mRNA in MS1 cells transfected with different combination of miRNAs based on qPCR (*P<0.05, compared with miRNA negative control). (e) The protein level of VEGFR2 in MS1 cells transfected with two combination of miRNAs based on western blotting. (f) Endothelial cell tube formation assay showed interference of network assembly of MS1 cells on pre-solidified Matrigel. Scale bar 500 μm (miRNA NC: Transfection of miRNA negative control; miRNAs: Transfection of miR (16-5p/322-5p/497-5p/); miRNAs+inhibitors: Transfection of miR (16-5p/322-5p/497-5p/17-5p) and inhibitors. *P<0.05).
Figure 8
Figure 8
miRNAs inhibitors rescued the effect of exosomes inhibition on VEGFR2 expression and tube formation in endothelial cells. (a) The relative expression of VEGFR2 mRNA in MS1 cells co-cultured with different groups exosomes and transfected with miRNAs inhibitors based on qPCR (***P<0.01). (b) The protein level of VEGFR2 in MS1 cells co-culture with Py or Py+LLC exosomes and miRNAs inhibitors based on western blotting. (c) Endothelial cell tube formation assay showed interference of network assembly of MS1 cells on presolidified Matrigel. Scale bar 500 μm (miRIn: Transfection of miR (16-5p/322-5p/497-5p/17-5p) inhibitors. ***P<0.01).
Figure 9
Figure 9
Schematic of plasma exosomes from plasmodium-infected mice inhibition of angiogenesis through miR(16/322/497/17) to target VEGFR2.

References

    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011; 144: 646–674.
    1. Bergers G, Benjamin LE. Tumorigenesis and the angiogenic switch. Nat Rev Cancer 2003; 3: 401–410.
    1. Hicklin DJ, Ellis LM. Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J Clin Oncol 2005; 23: 1011–1027.
    1. Ma W, Xu M, Liu Y, Liu H, Huang J, Zhu Y et al. Safety profile of combined therapy inhibiting EFGR and VEGF pathways in patients with advanced non-small-cell lung cancer: a meta-analysis of 15 phase II/III randomized trials. Int J Cancer 2015; 137: 409–419.
    1. Claesson-Welsh L, Welsh M. VEGFA and tumour angiogenesis. J Intern Med 2013; 273: 114–127.
    1. Nilsson I, Bahram F, Li X, Gualandi L, Koch S, Jarvius M et al. VEGF receptor 2/-3 heterodimers detected in situ by proximity ligation on angiogenic sprouts. EMBO J 2010; 29: 1377–1388.
    1. Ferrara N, Gerber H-P, LeCouter J. The biology of VEGF and its receptors. Nat Med 2003; 9: 669–676.
    1. Zachary I. VEGF signalling: integration and multi-tasking in endothelial cell biology. Biochem Soc T 2003; 31: 1171–1177.
    1. Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 2007; 9: 654–659.
    1. Nolte EN, Buermans HP, Waasdorp M, Stoorvogel W, Wauben MH, AC’t Hoen P. Deep sequencing of RNA from immune cell-derived vesicles uncovers the selective incorporation of small non-coding RNA biotypes with potential regulatory functions. Nucleic Acids Res 2012; 40: 9272–9285.
    1. Théry C. Exosomes: secreted vesicles and intercellular communications. F1000 biol rep 2011; 3: 130.
    1. Mantel PY, Marti M. The role of extracellular vesicles in Plasmodium and other protozoan parasites. Cell Microbiol 2014; 16: 344–354.
    1. Lener T, Gimona M, Aigner L, Börger V, Buzas E, Camussi G et al. Applying extracellular vesicles based therapeutics in clinical trials–an ISEV position paper. J extracell vesicles 2015; 4: 30087.
    1. Aline F, Bout D, Amigorena S, Roingeard P, Dimier-Poisson I. Toxoplasma gondii antigen-pulsed-dendritic cell-derived exosomes induce a protective immune response against T. gondii infection. Infect Immun 2004; 72: 4127–4137.
    1. Nantakomol D, Dondorp AM, Krudsood S, Udomsangpetch R, Pattanapanyasat K, Combes V et al. Circulating red cell-derived microparticles in human malaria. J Infect Dis 2011; 203: 700–706.
    1. Campos F, Franklin BS, Teixeira-Carvalho Filho A, Fontes C, Brito C, Carvalho L. Augmented plasma microparticles during acute Plasmodium vivax infection. Malaria J 2010; 9: 1475–2875.
    1. Martin-Jaular L, Nakayasu ES, Ferrer M, Almeida IC, Del Portillo HA. Exosomes from Plasmodium yoelii-infected reticulocytes protect mice from lethal infections. PLoS ONE 2011; 6: e26588.
    1. Chen L, He Z, Qin L, Li Q, Shi X, Zhao S et al. Antitumor effect of malaria parasite infection in a murine Lewis lung cancer model through induction of innate and adaptive immunity. PLoS ONE 2011; 6: e24407.
    1. Leong AS, Cooper K, Leong FJW. Manual of diagnostic antibodies for immunohistology. Cambridge University Press, 2003.
    1. Kerbel RS. Tumor angiogenesis. New Engl J Med 2008; 358: 2039–2049.
    1. Hasan MR, Ho SH, Owen DA, Tai IT. Inhibition of VEGF induces cellular senescence in colorectal cancer cells. Int J Cancer 2011; 129: 2115–2123.
    1. Combes V, Taylor TE, Juhan-Vague I, Mège J-L, Mwenechanya J, Tembo M et al. Circulating endothelial microparticles in malawian children with severe falciparum malaria complicated with coma. Jama 2004; 291: 2542–2544.
    1. Moxon CA, Chisala NV, Wassmer SC, Taylor TE, Seydel KB, Molyneux ME et al. Persistent endothelial activation and inflammation after Plasmodium falciparum infection in Malawian children. J Infect Dis 2014; 209: 610–615.
    1. Mfonkeu JP, Gouado I, Kuaté HF, Zambou O, Zollo PA, Grau GER et al. Elevated cell-specific microparticles are a biological marker for cerebral dysfunctions in human severe malaria. PLoS ONE 2010; 5: e13415–e13415.
    1. Marcilla A, Martin-Jaular L, Trelis M, de Menezes-Neto A, Osuna A, Bernal D et al. Extracellular vesicles in parasitic diseases. J extracell vesicles 2014; 3: 25040.
    1. Meckes DG, Raab-Traub N. Microvesicles and viral infection. J Virol 2011; 85: 12844–12854.
    1. Kulp A, Kuehn MJ. Biological functions and biogenesis of secreted bacterial outer membrane vesicles. Annu Rev Microbiol 2010; 64: 163.
    1. Ye S-b, Li Z-L, Luo D-h, Huang B-j, Chen Y-S, Zhang X-s et al. Tumor-derived exosomes promote tumor progression and T-cell dysfunction through the regulation of enriched exosomal microRNAs in human nasopharyngeal carcinoma. Oncotarget 2014; 5: 5439.
    1. Mantel PY, Hoang AN, Goldowitz I, Potashnikova D, Hamza B, Vorobjev I et al. Malaria-infected erythrocyte-derived microvesicles mediate cellular communication within the parasite population and with the host immune system. Cell Host Microbe 2013; 13: 521–534.
    1. Tang K, Zhang Y, Zhang H, Xu P, Liu J, Ma J et al. Delivery of chemotherapeutic drugs in tumour cell-derived microparticles. Nat Commun 2012; 3: 1282.
    1. Katakowski M, Buller B, Zheng X, Lu Y, Rogers T, Osobamiro O et al. Exosomes from marrow stromal cells expressing miR-146b inhibit glioma growth. Cancer Lett 2013; 335: 201–204.
    1. Roberts NJ, Zhang L, Janku F, Collins A, Bai R-Y, Staedtke V et al. Intratumoral injection of Clostridium novyi-NT spores induces antitumor responses. Sci Transl Med 2014; 6: 249ra111–249ra111.
    1. Lee S, Chen TT, Barber CL, Jordan MC, Murdock J, Desai S et al. Autocrine VEGF signaling is required for vascular homeostasis. Cell 2007; 130: 691–703.
    1. Yelamanchili SV, Lamberty BG, Rennard DA, Morsey BM, Hochfelder CG, Meays BM et al. MiR-21 in extracellular vesicles leads to neurotoxicity via TLR7 signaling in SIV neurological disease. Plos Pathog 2015; 11: e1005032.
    1. Zhou W, Fong MY, Min Y, Somlo G, Liu L, Palomares MR et al. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer cell 2014; 25: 501–515.
    1. Squadrito ML, Baer C, Burdet F, Maderna C, Gilfillan GD, Lyle R et al. Endogenous RNAs modulate microRNA sorting to exosomes and transfer to acceptor cells. Cell Rep 2014; 8: 1432–1446.
    1. Regev-Rudzki N, Wilson DW, Carvalho TG, Sisquella X, Coleman BM, Rug M et al. Cell-cell communication between malaria-infected red blood cells via exosome-like vesicles. Cell 2013; 153: 1120–1133.
    1. Mantel P-Y, Hjelmqvist D, Walch M, Kharoubi-Hess S, Nilsson S, Ravel D et al. Infected erythrocyte-derived extracellular vesicles alter vascular function via regulatory Ago2-miRNA complexes in malaria. Nat Commun 2016; 7: 12727.
    1. Calin GA, Cimmino A, Fabbri M, Ferracin M, Wojcik SE, Shimizu M et al. MiR-15a and miR-16-1 cluster functions in human leukemia. Proc Natl Acad Sci USA 2008; 105: 5166–5171.
    1. Bonci D, Coppola V, Musumeci M, Addario A, Giuffrida R, Memeo L et al. The miR-15a–miR-16-1 cluster controls prostate cancer by targeting multiple oncogenic activities. Nat Med 2008; 14: 1271–1277.
    1. Liu Q, Fu H, Sun F, Zhang H, Tie Y, Zhu J et al. miR-16 family induces cell cycle arrest by regulating multiple cell cycle genes. Nucleic Acids Res 2008; 36: 5391–5404.
    1. Finnerty JR, Wang W-X, Hébert SS, Wilfred BR, Mao G, Nelson PT. The miR-15/107 group of microRNA genes: evolutionary biology, cellular functions, and roles in human diseases. J Mol Biol 2010; 402: 491–509.
    1. Goretti E, Rolland-Turner M, Léonard F, Zhang L, Wagner DR, Devaux Y. MicroRNA-16 affects key functions of human endothelial progenitor cells. J Leukocyte Biol 2013; 93: 645–655.
    1. Doebele C, Bonauer A, Fischer A, Scholz A, Reiss Y, Urbich C et al. Members of the microRNA-17-92 cluster exhibit a cell-intrinsic antiangiogenic function in endothelial cells. Blood 2010; 115: 4944–4950.
    1. Ventura A, Young AG, Winslow MM, Lintault L, Meissner A, Erkeland SJ et al. Targeted deletion reveals essential and overlapping functions of the miR-17∼92 family of miRNA clusters. Cell 2008; 132: 875–886.

Source: PubMed

3
Se inscrever