Immune Netw.  2012 Jun;12(3):104-112. 10.4110/in.2012.12.3.104.

Induction of Functional Changes of Dendritic Cells by Silica Nanoparticles

Affiliations
  • 1Department of Biological Science and the Research Center for Women's Disease, Sookmyung Women's University, Seoul 140-742, Korea. jslim@sookmyung.ac.kr

Abstract

Silica is one of the most abundant compounds found in nature. Immoderate exposure to crystalline silica has been linked to pulmonary disease and crystalline silica has been classified as a Group I carcinogen. Ultrafine (diameter <100 nm) silica particles may have different toxicological properties compared to larger particles. We evaluated the effect of ultrafine silica nanoparticles on mouse bone marrow-derived dendritic cells (BMDC) and murine dendritic cell line, DC2.4. The exposure of dendritic cells (DCs) to ultrafine silica nanoparticles showed a decrease in cell viability and an induction of cell death in size- and concentration-dependent manners. In addition, in order to examine the phenotypic changes of DCs following co-culture with silica nanoparticles, we added each sized-silica nanoparticle along with GM-CSF and IL-4 during and after DC differentiation. Expression of CD11c, a typical DC marker, and multiple surface molecules such as CD54, CD80, CD86, MHC class II, was changed by silica nanoparticles in a size-dependent manner. We also found that silica nanoparticles affect inflammatory response in DCs in vitro and in vivo. Finally, we found that p38 and NF-kappaB activation may be critical for the inflammatory response by silica nanoparticles. Our data demonstrate that ultrafine silica nanoparticles have cytotoxic effects on dendritic cells and immune modulation effects in vitro and in vivo.

Keyword

Silica nanoparticles; Dendritic cells; Apoptosis; Inflammatory response

MeSH Terms

Animals
Apoptosis
Cell Death
Cell Survival
Coculture Techniques
Crystallins
Dendritic Cells
Granulocyte-Macrophage Colony-Stimulating Factor
Interleukin-4
Lung Diseases
Mice
Nanoparticles
NF-kappa B
Silicon Dioxide
Silicones
Crystallins
Granulocyte-Macrophage Colony-Stimulating Factor
Interleukin-4
NF-kappa B
Silicon Dioxide
Silicones

Figure

  • Figure 1 Induction of dendritic cell death by silica nanoparticles. For determining cell viability of DC2.4 cells after treatment with silica nanoparticles, cells were treated with silica nanoparticles for 24 h and 48 h. Cell viability was determined using MTT assay (A) and trypan blue staining (B). Cells were treated with silica nanoparticles (40 µg/ml) for 24 h and 7-AAD (PerCP) and annexin V-FITC staining was used to analyze the DC2.4 cell death. The percentages of stained/unstained cells are shown in each quadrant (C). The total sum of annexin V-positive cells was quantified (D). Results represent the mean±SD from triplicates. *p<0.05, **p<0.01, ***p<0.001 for one-way ANOVA.

  • Figure 2 Silica nanoparticles affect the expression of surface antigens on dendritic cells. Bone marrow-derived DCs were generated by culturing bone marrow cells with 10 ng/ml of GM-CSF and 10 ng/ml of IL-4 for 6 days. Thereafter, cells were co-cultured with silica nanoparticles (40 µg/ml) for 24 h (A). On the other hand, bone marrow-derived cells were cultured with 10 ng/ml of GM-CSF, 10 ng/ml of IL-4, and silica nanoparticles (10 µg/ml) for 7 days (B). Cells were allowed to react with appropriate antibodies at 4℃ for 30 min for the detection of CD11c, CD54, CD80, CD86 and MHC class II. Cells were analyzed using a FACSCanto™II. The data are represented as relative Mean Fluorescence Intensity (MFI).

  • Figure 3 Silica nanoparticles induce TNF-α production in dendritic cells. DC2.4 cells (A) and bone marrow-derived dendritic cells (B, D, and F) were exposed to silica nanoparticles (40 µg/ml) for 24 h. Then, bone marrow-derived cells were cultured with 10 ng/ml of GM-CSF, 10 ng/ml of IL-4, and 10 µg/ml of silica nanoparticles for 7 days (C, E, and G). Cells were harvested for RNA preparation. Transcriptional levels of cytokines were detected using RT-PCR (A~C). The supernatants of BMDCs were collected for 24 h. The levels of TNF-α (D and E) and IL-12p70 (F and G) in culture supernatants were determined by ELISA. Data represent the mean±SD of duplicates. *p<0.05, **p<0.01 for one-way ANOVA.

  • Figure 4 Silica nanoparticles elicit inflammatory responses in vivo. C57BL/6 mice were injected subcutaneously with 700 µl liquid matrigel containing silica nanoparticles (10 mg/kg). After 10 days, gels were excised (A) and hemoglobin in matrigel plugs was determined using Drabkin's reagent (B). BMDCs were exposed to silica nanoparticles (40 µg/ml) for 24 h (C and D). In addition, bone marrow-derived cells were cultured with 10 ng/ml of GM-CSF, 10 ng/ml of IL-4, and 10 µg/ml of silica nanoparticles for 7 days (E and F). The culture supernatants were collected for last 24 h and concentrated with centricon centrifugal filter devices. A mixture of 500 µl liquid matrigel and 200 µl concentrated supernatant was injected to C57BL/6 mice subcutaneously. After 9~10 days, gels were excised (C and E) and hemoglobin in matrigel plugs was determined (D and F). DC 2.4 cells were treated with silica nanoparticles (40 µg/ml) for 24 h. DC2.4 cells treated with silica nanoparticles were isolated by a density gradient centrifugation on ficoll to remove contamination of silica nanoparticles. C57BL/6 mice were injected subcutaneously with 700 µl liquid matrigel containing 5×105 DC2.4 cells. After 10 days, gels were excised and matrigel plugs were stained with hematoxylin & eosin (H&E) (G). Results represent the mean±SD from duplicates. *p<0.05 for one-way ANOVA.

  • Figure 5 Phosphorylation of p38 MAPK is essentially associated with the induction of in vivo inflammatory responses by nanoparticles. DC2.4 cells were co-cultured with 40 µg/ml of silica nanoparticles for 6 h. Equal amounts of whole lysates were subjected to electrophoresis on a SDS-PAGE and Western blot analysis was performed using specific antibodies, respectively.


Reference

1. Chen M, von Mikecz A. Formation of nucleoplasmic protein aggregates impairs nuclear function in response to SiO2 nanoparticles. Exp Cell Res. 2005. 305:51–62.
Article
2. Pan Z, Lee W, Slutsky L, Clark RA, Pernodet N, Rafailovich MH. Adverse effects of titanium dioxide nanoparticles on human dermal fibroblasts and how to protect cells. Small. 2009. 5:511–520.
Article
3. Foley S, Crowley C, Smaihi M, Bonfils C, Erlanger BF, Seta P, Larroque C. Cellular localisation of a water-soluble fullerene derivative. Biochem Biophys Res Commun. 2002. 294:116–119.
Article
4. Oberdörster E. Manufactured nanomaterials (fullerenes, C60) induce oxidative stress in the brain of juvenile largemouth bass. Environ Health Perspect. 2004. 112:1058–1062.
Article
5. Vallhov H, Gabrielsson S, Strømme M, Scheynius A, Garcia-Bennett AE. Mesoporous silica particles induce size dependent effects on human dendritic cells. Nano Lett. 2007. 7:3576–3582.
Article
6. Cho WS, Choi M, Han BS, Cho M, Oh J, Park K, Kim SJ, Kim SH, Jeong J. Inflammatory mediators induced by intratracheal instillation of ultrafine amorphous silica particles. Toxicol Lett. 2007. 175:24–33.
Article
7. Huang DM, Chung TH, Hung Y, Lu F, Wu SH, Mou CY, Yao M, Chen YC. Internalization of mesoporous silica nanoparticles induces transient but not sufficient osteogenic signals in human mesenchymal stem cells. Toxicol Appl Pharmacol. 2008. 231:208–215.
Article
8. Wang JJ, Sanderson BJ, Wang H. Cytotoxicity and genotoxicity of ultrafine crystalline SiO2 particulate in cultured human lymphoblastoid cells. Environ Mol Mutagen. 2007. 48:151–157.
Article
9. 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.
Article
10. Fubini B, Hubbard A. Reactive oxygen species (ROS) and reactive nitrogen species (RNS) generation by silica in inflammation and fibrosis. Free Radic Biol Med. 2003. 34:1507–1516.
Article
11. Warheit DB, McHugh TA, Hartsky MA. Differential pulmonary responses in rats inhaling crystalline, colloidal or amorphous silica dusts. Scand J Work Environ Health. 1995. 21:Suppl 2. 19–21.
12. Leigh J, Wang H, Bonin A, Peters M, Ruan X. Silica-induced apoptosis in alveolar and granulomatous cells in vivo. Environ Health Perspect. 1997. 105:Suppl 5. 1241–1245.
13. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998. 392:245–252.
Article
14. Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ, Pulendran B, Palucka K. Immunobiology of dendritic cells. Annu Rev Immunol. 2000. 18:767–811.
Article
15. Blanco P, Palucka AK, Pascual V, Banchereau J. Dendritic cells and cytokines in human inflammatory and autoimmune diseases. Cytokine Growth Factor Rev. 2008. 19:41–52.
Article
16. Kang K, Kim H, Kim KI, Yang Y, Yoon DY, Kim JH, Ryu JH, Noh EJ, Jeon SD, Lim JS. SK-126, a synthetic compound, regulates the production of inflammatory cytokines induced by LPS in antigen-presenting cells. Biochem Pharmacol. 2008. 75:1054–1064.
Article
17. Heath WR, Belz GT, Behrens GM, Smith CM, Forehan SP, Parish IA, Davey GM, Wilson NS, Carbone FR, Villadangos JA. Cross-presentation, dendritic cell subsets, and the generation of immunity to cellular antigens. Immunol Rev. 2004. 199:9–26.
Article
18. Reis e Sousa C. Dendritic cells in a mature age. Nat Rev Immunol. 2006. 6:476–483.
Article
19. Shen Z, Reznikoff G, Dranoff G, Rock KL. Cloned dendritic cells can present exogenous antigens on both MHC class I and class II molecules. J Immunol. 1997. 158:2723–2730.
20. Inaba K, Inaba M, Romani N, Aya H, Deguchi M, Ikehara S, Muramatsu S, Steinman RM. Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor. J Exp Med. 1992. 176:1693–1702.
Article
21. Wörle-Knirsch JM, Pulskamp K, Krug HF. Oops they did it again! Carbon nanotubes hoax scientists in viability assays. Nano Lett. 2006. 6:1261–1268.
Article
22. Laaksonen T, Santos H, Vihola H, Salonen J, Riikonen J, Heikkilä T, Peltonen L, Kumar N, Murzin DY, Lehto VP, Hirvonen J. Failure of MTT as a toxicity testing agent for mesoporous silicon microparticles. Chem Res Toxicol. 2007. 20:1913–1918.
Article
23. Kowalczyk DW, Wlazlo AP, Blaszczyk-Thurin M, Xiang ZQ, Giles-Davis W, Ertl HC. A method that allows easy characterization of tumor-infiltrating lymphocytes. J Immunol Methods. 2001. 253:163–175.
Article
24. Curiel TJ, Cheng P, Mottram P, Alvarez X, Moons L, Evdemon-Hogan M, Wei S, Zou L, Kryczek I, Hoyle G, Lackner A, Carmeliet P, Zou W. Dendritic cell subsets differentially regulate angiogenesis in human ovarian cancer. Cancer Res. 2004. 64:5535–5538.
Article
25. Saudemont A, Jouy N, Hetuin D, Quesnel B. NK cells that are activated by CXCL10 can kill dormant tumor cells that resist CTL-mediated lysis and can express B7-H1 that stimulates T cells. Blood. 2005. 105:2428–2435.
Article
26. Kang K, Lim DH, Choi IH, Kang T, Lee K, Moon EY, Yang Y, Lee MS, Lim JS. Vascular tube formation and angiogenesis induced by polyvinylpyrrolidone-coated silver nanoparticles. Toxicol Lett. 2011. 205:227–234.
Article
27. Hansen SF, Michelson ES, Kamper A, Borling P, Stuer-Lauridsen F, Baun A. Categorization framework to aid exposure assessment of nanomaterials in consumer products. Ecotoxicology. 2008. 17:438–447.
Article
28. Passagne I, Morille M, Rousset M, Pujalté I, L'azou B. Implication of oxidative stress in size-dependent toxicity of silica nanoparticles in kidney cells. Toxicology. 2012. 299:112–124.
Article
29. Napierska D, Thomassen LC, Rabolli V, Lison D, Gonzalez L, Kirsch-Volders M, Martens JA, Hoet PH. Size-dependent cytotoxicity of monodisperse silica nanoparticles in human endothelial cells. Small. 2009. 5:846–853.
Article
30. Waters KM, Masiello LM, Zangar RC, Tarasevich BJ, Karin NJ, Quesenberry RD, Bandyopadhyay S, Teeguarden JG, Pounds JG, Thrall BD. Macrophage responses to silica nanoparticles are highly conserved across particle sizes. Toxicol Sci. 2009. 107:553–569.
Article
31. Lin W, Huang YW, Zhou XD, Ma Y. In vitro toxicity of silica nanoparticles in human lung cancer cells. Toxicol Appl Pharmacol. 2006. 217:252–259.
Article
32. Cha KE, Myung H. Cytotoxic effects of nanoparticles assessed in vitro and in vivo. J Microbiol Biotechnol. 2007. 17:1573–1578.
33. Choi SJ, Oh JM, Choy JH. Toxicological effects of inorganic nanoparticles on human lung cancer A549 cells. J Inorg Biochem. 2009. 103:463–471.
Article
34. Carlson C, Hussain SM, Schrand AM, Braydich-Stolle LK, Hess KL, Jones RL, Schlager JJ. Unique cellular interaction of silver nanoparticles: size-dependent generation of reactive oxygen species. J Phys Chem B. 2008. 112:13608–13619.
Article
35. Park EJ, Yi J, Chung KH, Ryu DY, Choi J, Park K. Oxidative stress and apoptosis induced by titanium dioxide nanoparticles in cultured BEAS-2B cells. Toxicol Lett. 2008. 180:222–229.
Article
36. Herzog E, Byrne HJ, Casey A, Davoren M, Lenz AG, Maier KL, Duschl A, Oostingh GJ. SWCNT suppress inflammatory mediator responses in human lung epithelium in vitro. Toxicol Appl Pharmacol. 2009. 234:378–390.
Article
Full Text Links
  • IN
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr