Nano-sized and micro-sized polystyrene particles affect phagocyte function

B Prietl, C Meindl, E Roblegg, T R Pieber, G Lanzer, E Fröhlich, B Prietl, C Meindl, E Roblegg, T R Pieber, G Lanzer, E Fröhlich

Abstract

Adverse effect of nanoparticles may include impairment of phagocyte function. To identify the effect of nanoparticle size on uptake, cytotoxicity, chemotaxis, cytokine secretion, phagocytosis, oxidative burst, nitric oxide production and myeloperoxidase release, leukocytes isolated from human peripheral blood, monocytes and macrophages were studied. Carboxyl polystyrene (CPS) particles in sizes between 20 and 1,000 nm served as model particles. Twenty nanometers CPS particles were taken up passively, while larger CPS particles entered cells actively and passively. Twenty nanometers CPS were cytotoxic to all phagocytes, ≥500 nm CPS particles only to macrophages. Twenty nanometers CPS particles stimulated IL-8 secretion in human monocytes and induced oxidative burst in monocytes. Five hundred nanometers and 1,000 nm CPS particles stimulated IL-6 and IL-8 secretion in monocytes and macrophages, chemotaxis towards a chemotactic stimulus of monocytes and phagocytosis of bacteria by macrophages and provoked an oxidative burst of granulocytes. At very high concentrations, CPS particles of 20 and 500 nm stimulated myeloperoxidase release of granulocytes and nitric oxide generation in macrophages. Cytotoxic effect could contribute to some of the observed effects. In the absence of cytotoxicity, 500 and 1,000 nm CPS particles appear to influence phagocyte function to a greater extent than particles in other sizes.

Figures

Fig. 1
Fig. 1
Uptake of fluorescently labelled carboxyl polystyrene particles (FluoSpheres®, FS) in PBMCs exposed in DMEM. After short incubation at 37 °C, lymphocytes (CD3-immunoreactive) take up 20 nm FS but not larger particles. Granulocytes (CD13-immunoreactive) and monocytes (CD14-immunoreactive) also ingest 200 and 500 nm FS after this time. Uptake of ≥200 nm FS is inhibited by low temperature. After 24 h incubation, lymphocytes also contain 500 nm FS. Asterisks indicate FS taken up by platelets. Scale bar 10 μm
Fig. 2
Fig. 2
Viability of human monocytic U937 and THP-1 cells, differentiated THP-1 cells (THPdiff) and murine DMBM-2 macrophages after incubation with carboxyl polystyrene particles (CPS particles) of ≤200 nm (a) and ≥500 nm (b) for 24 h exposed in DMEM (n=4)
Fig. 3
Fig. 3
Viability of DMBM-2 macrophages (a) and THP-1 cells (b) dependent on particles/cell (n=4). CPS particles decrease viability of monocytic THP-1 cells at lower numbers than viability of DMBM-2 macrophages
Fig. 4
Fig. 4
Chemotaxis of U937 cells towards a positive chemotactic stimulus (PMA) after incubation for 30 min with plasma-coated CPS particles (n=3). Significant changes to the solvent-treated controls (p<0.05) and between different particles linked by brackets are indicated by asterisk. A hatch indicates significant difference of one particle to the others
Fig. 5
Fig. 5
Release of cytokines IL-6 (a) and IL-8 (b) from monocytic THP-1 cells upon stimulation with 20 μg/ml CPS particles suspended in DMEM for 24 h (n=6). THP-1 cells were also assessed after differentiation with PMA and releases were normalised to solvent-treated controls as 1. a IL-6 secretion was increased by incubations with CPS ≥500 nm. b 20 nm CPS stimulated IL-8 secretion in differentiated and monocytic THP-1; 100 ng/ml LPS served as positive control. Significant changes to the solvent-treated controls (p<0.05) and between different particles linked by brackets are indicated by asterisk
Fig. 6
Fig. 6
Phagocytosis of bacteria by DMBM-2 macrophages after pre-incubation with CPS particles suspended in DMEM (n=3). Twenty nanometers CPS particles inhibited phagocytosis, while 1,000 and 500 nm CPS particles stimulated the uptake. Data are normalised (100 %) to the uptake of indicator particles in medium. Significant changes to the solvent-treated controls (p<0.05) are indicated by asterisk
Fig. 7
Fig. 7
Generation of superoxide in monocytes (a) and granulocytes (b) in human blood samples exposed to 25 μg/ml CPS particles suspended in PBS (n=4). E. coli provided in the assay kit served as positive control (PC) and PBS as negative control (NC). Data are normalised to the generation of superoxide in cells exposed to buffer. a Uncoated and plasma-coated 20 nm CPS particles increase the production of superoxide by monocytes while 1,000 nm CPS particles display only a marginal effect. b In granulocytes, plasma-coated 1,000 nm CPS particles increase the production of superoxide while 20 nm CPS particles display only a marginal effect. Significant changes to the solvent-treated controls (p<0.05) are indicated by asterisk. u uncoated, c plasma coated

References

    1. Allermann L, Poulsen OM. Interleukin-8 secretion from monocytic cell lines for evaluation of the inflammatory potential of organic dust. Environ Res. 2002;88:188–98.
    1. Bernard L, Vaudaux P, Huggler E, Stern R, Frehel C, Francois P, et al. Inactivation of a subpopulation of human neutrophils by exposure to ultrahigh-molecular-weight polyethylene wear debris. FEMS Immunol Med Microbiol. 2007;49:425–32.
    1. Catelas I, Huk OL, Petit A, Zukor DJ, Marchand R, Yahia L. Flow cytometric analysis of macrophage response to ceramic and polyethylene particles: effects of size, concentration, and composition. J Biomed Mater Res. 1998;41:600–7.
    1. Champion JA, Mitragotri S. Shape induced inhibition of phagocytosis of polymer particles. Pharm Res. 2009;26:244–9.
    1. Fröhlich E, Samberger C, Kueznik T, Absenger M, Roblegg E, Zimmer A, et al. Cytotoxicity of nanoparticles independent from oxidative stress. J Toxicol Sci. 2009;34:363–75.
    1. Fröhlich E, Meindl C, Roblegg E, Ebner B, Absenger M, Pieber TR. Action of polystyrene nanoparticles of different sizes on lysosomal function and integrity. Part Fibre Toxicol. 2012a;9:26.
    1. Fröhlich E, Meindl C, Roblegg E, Griesbacher A, Pieber TR. Cytotoxicity of nanoparticles is influenced by size, proliferation and embryonic origin of the cells used for testing. Nanotoxicology. 2012b;6:424–3.
    1. Fröhlich E, Bonstingl G, Hofler A, Meindl C, Leitinger G, Pieber TR, et al. Comparison of two in vitro systems to assess cellular effects of nanoparticles-containing aerosols. Toxicol in vitro. 2013;27:409–17.
    1. Frokjaer J, Deleuran B, Lind M, Overgaard S, Soballe K, Bunger C. Polyethylene particles stimulate monocyte chemotactic and activating factor production in synovial mononuclear cells in vivo. An immunohistochemical study in rabbits. Acta Orthop Scand. 1995;66:303–7.
    1. Garrett R, Wilksch J, Vernon-Roberts B. Effects of cobalt-chrome alloy wear particles on the morphology, viability and phagocytic activity of murine macrophages in vitro. Aust J Exp Biol Med Sci. 1983;61(Pt 3):355–69.
    1. Gosens I, Post JA, de la Fonteyne LJ, Jansen EH, Geus JW, Cassee FR, et al. Impact of agglomeration state of nano- and submicron sized gold particles on pulmonary inflammation. Part Fibre Toxicol. 2010;7:37.
    1. Hedenborg M. Titanium dioxide induced chemiluminescence of human polymorphonuclear leukocytes. Int Arch Occup Environ Health. 1988;61:1–6.
    1. Heng BC, Zhao X, Tan EC, Khamis N, Assodani A, Xiong S, et al. Evaluation of the cytotoxic and inflammatory potential of differentially shaped zinc oxide nanoparticles. Arch Toxicol. 2011;85:1517–28.
    1. Hirst SM, Karakoti AS, Tyler RD, Sriranganathan N, Seal S, Reilly CM. Anti-inflammatory properties of cerium oxide nanoparticles. Small. 2009;5:2848–56.
    1. Hsiao JK, Chu HH, Wang YH, Lai CW, Chou PT, Hsieh ST, et al. Macrophage physiological function after superparamagnetic iron oxide labeling. NMR Biomed. 2008;21:820–9.
    1. Hutter E, Boridy S, Labrecque S, Lalancette-Hebert M, Kriz J, Winnik FM, et al. Microglial response to gold nanoparticles. ACS nano. 2010;4:2595–606.
    1. Inoue K, Takano H, Yanagisawa R, Koike E, Shimada A. Size effects of latex nanomaterials on lung inflammation in mice. Toxicol Appl Pharmacol. 2009;234:68–76.
    1. Jones BG, Dickinson PA, Gumbleton M, Kellaway IW. The inhibition of phagocytosis of respirable microspheres by alveolar and peritoneal macrophages. Int J Pharm. 2002;236:65–79.
    1. Jovanovic B, Anastasova L, Rowe EW, Zhang Y, Clapp AR, Palic D. Effects of nanosized titanium dioxide on innate immune system of fathead minnow (Pimephales promelas Rafinesque, 1820) Ecotoxicol Environ Saf. 2011;74:675–83.
    1. Kumazawa R, Watari F, Takashi N, Tanimura Y, Uo M, Totsuka Y. Effects of Ti ions and particles on neutrophil function and morphology. Biomaterials. 2002;23:3757–64.
    1. Lesniak A, Fenaroli F, Monopoli MP, Aberg C, Dawson KA, Salvati A. Effects of the presence or absence of a protein corona on silica nanoparticle uptake and impact on cells. ACS nano. 2012;6:5845–57.
    1. Lesniak A, Salvati A, Santos-Martinez MJ, Radomski MW, Dawson KA, Aberg C. Nanoparticle adhesion to the cell membrane and its effect on nanoparticle uptake efficiency. J Am Chem Soc. 2013;135:1438–44.
    1. Liu R, Yin LH, Pu YP, Li YH, Zhang XQ, Liang GY, et al. The immune toxicity of titanium dioxide on primary pulmonary alveolar macrophages relies on their surface area and crystal structure. J Nanosci Nanotechnol. 2010;10:8491–9.
    1. Lundqvist M, Stigler J, Elia G, Lynch I, Cedervall T, Dawson KA. Nanoparticle size and surface properties determine the protein corona with possible implications for biological impacts. Proc Natl Acad Sci U S A. 2008;105:14265–70.
    1. Mainardes RM, Gremiao MP, Brunetti IL, da Fonseca LM, Khalil NM. Zidovudine-loaded PLA and PLA-PEG blend nanoparticles: influence of polymer type on phagocytic uptake by polymorphonuclear cells. J Pharm Sci. 2009;98:257–67.
    1. Nie S. Understanding and overcoming major barriers in cancer nanomedicine. Nanomedicine: Nanotech Biol Med. 2010;5:523–8.
    1. Nishimori H, Kondoh M, Isoda K, Tsunoda S, Tsutsumi Y, Yagi K. Silica nanoparticles as hepatotoxicants. Eur J Pharm Biopharm. 2009;72:496–501.
    1. Olivier V, Duval JL, Hindie M, Pouletaut P, Nagel MD. Comparative particle-induced cytotoxicity toward macrophages and fibroblasts. Cell Biol Toxicol. 2003;19:145–59.
    1. Papatheofanis FJ, Barmada R. Polymorphonuclear leukocyte degranulation with exposure to polymethylmethacrylate nanoparticles. J Biomed Mater Res. 1991;25:761–71.
    1. Park EJ, Park K. Oxidative stress and pro-inflammatory responses induced by silica nanoparticles in vivo and in vitro. Toxicol Lett. 2009;184:18–25.
    1. Renwick LC, Donaldson K, Clouter A. Impairment of alveolar macrophage phagocytosis by ultrafine particles. Toxicol Appl Pharmacol. 2001;172:119–27.
    1. Roblegg E, Fröhlich E, Meindl C, Teubl B, Zaversky M, Zimmer A. Evaluation of a physiological in vitro system to study the transport of nanoparticles through the buccal mucosa. Nanotoxicology. 2012;6:399–413.
    1. Rothen-Rutishauser BM, Schurch S, Haenni B, Kapp N, Gehr P. Interaction of fine particles and nanoparticles with red blood cells visualized with advanced microscopic techniques. Environ Sci Technol. 2006;40:4353–9.
    1. Sandberg W, Låg M, Holme J, Friede B, Gualtieri M, Kruszewski M, et al. Comparison of non-crystalline silica nanoparticles in IL-1ß release from macrophages. Part Fibre Toxicol. 2012;9:32.
    1. Scheel J, Weimans S, Thiemann A, Heisler E, Hermann M. Exposure of the murine RAW 264.7 macrophage cell line to hydroxyapatite dispersions of various composition and morphology: assessment of cytotoxicity, activation and stress response. Toxicol in vitro. 2009;23:531–8.
    1. Scherbart AM, Langer J, Bushmelev A, van Berlo D, Haberzettl P, van Schooten FJ, et al. Contrasting macrophage activation by fine and ultrafine titanium dioxide particles is associated with different uptake mechanisms. Part Fibre Toxicol. 2011;8:31.
    1. Segat D, Tavano R, Donini M, Selvestrel F, Rio-Echevarria I, Rojnik M, et al. Proinflammatory effects of bare and PEGylated ORMOSIL-, PLGA- and SUV-NPs on monocytes and PMNs and their modulation by f-MLP. Nanomedicine: Nanotech Biol Med. 2011;6:1027–46.
    1. Shavandi Z, Ghazanfari T, Moghaddam KN. In vitro toxicity of silver nanoparticles on murine peritoneal macrophages. Immunopharmacol Immunotoxicol. 2011;33:135–40.
    1. Shwe TT, Yamamoto S, Kakeyama M, Kobayashi T, Fujimaki H. Effect of intratracheal instillation of ultrafine carbon black on proinflammatory cytokine and chemokine release and mRNA expression in lung and lymph nodes of mice. Toxicol Appl Pharmacol. 2005;209:51–61.
    1. Tenzer S, Docter D, Kuharev J, Musyanovych A, Fetz V, Hecht R, et al. Rapid formation of plasma protein corona critically affects nanoparticle pathophysiology. Nat Nanotechnol. 2013;8:772–81.
    1. Vesnina LE, Mamontova TV, Mikitiuk MV, Kutsenko NL, Kutsenko LA, Bobrova NA, et al. Effect of fullerene C60 on functional activity of phagocytic cells. Eksp Klin Farmakol. 2011;74:26–9.
    1. Villiers C, Freitas H, Couderc R, Villiers MB, Marche P. Analysis of the toxicity of gold nano particles on the immune system: effect on dendritic cell functions. J Nanopart Res. 2010;12:55–60.
    1. Walter G, Santra S, Thattaliyath B, Grant S. (Super)paramagnetic nanoparticles: applications in noninvasive MR imaging of stem cell transfer. In: Bulte J, Modo M, editors. Fundamental biomedical technologies nanoparticles in biomedical imaging emerging technologies and applications. Springer; New York: 2008.
    1. Winter M, Beer HD, Hornung V, Kramer U, Schins RP, Forster I. Activation of the inflammasome by amorphous silica and TiO2 nanoparticles in murine dendritic cells. Nanotoxicology. 2011;5:326–40.
    1. Witasp E, Shvedova AA, Kagan VE, Fadeel B. Single-walled carbon nanotubes impair human macrophage engulfment of apoptotic cell corpses. Inhal Toxicol. 2009;21(Suppl 1):131–6.
    1. Yang EJ, Kim S, Kim JS, Choi IH. Inflammasome formation and IL-1beta release by human blood monocytes in response to silver nanoparticles. Biomaterials. 2012;33:6858–67.
    1. Yue H, Wei W, Yue Z, Lv P, Wang L, Ma G, et al. Particle size affects the cellular response in macrophages. Eur J Pharm Sci. 2010;41:650–7.

Source: PubMed

3
Tilaa