Immune Netw.  2011 Jun;11(3):163-168. 10.4110/in.2011.11.3.163.

Formulation and Characterization of Antigen-loaded PLGA Nanoparticles for Efficient Cross-priming of the Antigen

Affiliations
  • 1College of Pharmacy, Chungbuk National University, Cheongju 361-763, Korea. cklee@chungbuk.ac.kr
  • 2College of Pharmacy, ShamYook University, Seoul 139-742, Korea.

Abstract

BACKGROUND
Nanoparticles (NPs) prepared from biodegradable polymers, such as poly (D,L-lactic acid-co-glycolic acid) (PLGA), have been studied as vehicles for the delivery of antigens to phagocytes. This paper describes the preparation of antigen-loaded PLGA-NPs for efficient cross-priming.
METHODS
NPs containing a similar amount of ovalbumin (OVA) but different sizes were produced using a micromixer-based W/O/W solvent evaporation procedure, and the efficiency of the NPs to induce the cross-presentation of OVA peptides were examined in dendritic cells (DCs). Cellular uptake and biodistribution studies were performed using fluorescein isothiocyanate (FITC)-loaded NPs in mice.
RESULTS
The NPs in the range of 1.1~1.4microm in size were the most and almost equally efficient in inducing the cross-presentation of OVA peptides via H-2Kb molecules. Cellular uptake and biodistribution studies showed that opsonization of the NPs with mouse IgG greatly increased the percentage of FITC-positive cells in the spleen and lymph nodes. The major cell type of FITC-positive cells in the spleen was macrophages, whereas that of lymph nodes was DCs.
CONCLUSION
These results show that IgG-opsonized PLGA-NPs with a mean size of 1.1microm would be the choice of biodegradable carriers for the targeted-delivery of protein antigens for cross-priming in vivo.

Keyword

PLGA; Nanoparticle; Opsonization; Cross-priming

MeSH Terms

Animals
Cross-Priming
Dendritic Cells
Fluorescein
Immunoglobulin G
Isothiocyanates
Lactic Acid
Lymph Nodes
Macrophages
Mice
Nanoparticles
Ovalbumin
Ovum
Peptides
Phagocytes
Polyglycolic Acid
Polymers
Spleen
Fluorescein
Immunoglobulin G
Isothiocyanates
Lactic Acid
Ovalbumin
Peptides
Polyglycolic Acid
Polymers

Figure

  • Figure 1 Release profile of OVA from OVA-loaded NPs. (A) Scanning electron microscopy showing the surface morphology of NPs with a mean size of 1.1µm. (B) Four milligrams of the OVA-NPs were dispersed in 4 ml of PBS containing 0.02% sodium azide, and then were kept at 37℃, or 4℃. At the indicated days, the supernatants were collected after centrifugation, and then the protein contents were analyzed using a micro-bicinchoninic acid assay.

  • Figure 2 Effects of the size of NPs on the cross-presentation. DCs were incubated with NPs with different mean sizes for 2 h, washed with PBS, fixed with paraformaldehyde, and the amounts of OVA peptides presented on MHC class I molecules were assessed by a LacZ T cell activation assay using OVA specific CD8 T cell hybridomas, B3Z cells.

  • Figure 3 Time-kinetics for the cross-presentation of OVA peptides. DCs were incubated with soluble OVA (50µg/ml), OVA-loaded NPs (50µg/ml), IgG-opsonized OVA-loaded NPs (50µg/ml), or 1 nM synthetic OVA peptide (SIINFEKL) for 2 h. The DCs were then washed with PBS, and were cultured in a medium containing 10% fetal bovine serum. At the indicated time points, the DCs were fixed with paraformaldehyde, and the amounts of OVA peptides presented on MHC class I molecules were assessed by a LacZ T cell activation assay using OVA specific CD8 T cell hybridomas, B3Z cells.

  • Figure 4 The uptake and biodistribution of NPs injected into the mouse peritoneum. C57BL/6 mice were injected i.p. with 100µg of NPs containing both OVA and FITC. Mice were sacrificed 2 h later, and mononuclear cell suspensions were prepared from the spleens (A) and lymph nodes (popliteal, inguinal, mesenteric, and axillary) (B). The single cell suspensions were fixed with paraformaldehyde and analyzed for FITC-positive cells by flow cytometry.

  • Figure 5 Cell type analysis for the FITC-positive cells in the spleen and lymph nodes. C57BL/6 mice were injected i.p. with 100µg of NPs containing both OVA and FITC. The mice were sacrificed 2 h later, and mononuclear cell suspensions were prepared from the spleens (A) and lymph nodes (B), stained with phycoerythrin-labeled anti-mouse CD11b, CD11c, or Gr-1 monoclonal antibodies, and analyzed for FITC-positive cells by flow cytometry.


Cited by  2 articles

Induction of Potent Antigen-specific Cytotoxic T Cell Response by PLGA-nanoparticles Containing Antigen and TLR Agonist
Young-Ran Lee, Young-Hee Lee, Ki-Hyang Kim, Sun-A Im, Chong-Kil Lee
Immune Netw. 2013;13(1):30-33.    doi: 10.4110/in.2013.13.1.30.

Nanoliposomes of L-lysine-conjugated poly(aspartic acid) Increase the Generation and Function of Bone Marrow-derived Dendritic Cells
Sun-A Im, Ki-Hyang Kim, Hong-Geun Ji, Hyoung-Gyoung Yu, Sun-Ki Park, Chong-Kil Lee
Immune Netw. 2011;11(5):281-287.    doi: 10.4110/in.2011.11.5.281.


Reference

1. Bevan MJ. Cross-priming for a secondary cytotoxic response to minor H antigens with H-2 congenic cells which do not cross-react in the cytotoxic assay. J Exp Med. 1976. 143:1283–1288.
Article
2. Champion JA, Walker A, Mitragotri S. Role of particle size in phagocytosis of polymeric microspheres. Pharm Res. 2008. 25:1815–1821.
Article
3. Elamanchili P, Diwan M, Cao M, Samuel J. Characterization of poly(D,L-lactic-co-glycolic acid) based nanoparticulate system for enhanced delivery of antigens to dendritic cells. Vaccine. 2004. 22:2406–2412.
Article
4. Gerelchuluun T, Lee YH, Lee YR, Im SA, Song S, Park JS, Han K, Kim K, Lee CK. Dendritic cells process antigens encapsulated in a biodegradable polymer, poly(D,L-lactide-co-glycolide), via an alternate class I MHC processing pathway. Arch Pharm Res. 2007. 30:1440–1446.
Article
5. Harding CV, Song R. Phagocytic processing of exogenous particulate antigens by macrophages for presentation by class I MHC molecules. J Immunol. 1994. 153:4925–4933.
6. Heath WR, Carbone FR. Cross-presentation, dendritic cells, tolerance and immunity. Annu Rev Immunol. 2001. 19:47–64.
Article
7. Huang AY, Golumbek P, Ahmadzadeh M, Jaffee E, Pardoll D, Levitsky H. Role of bone marrow-derived cells in presenting MHC class I-restricted tumor antigens. Science. 1994. 264:961–965.
Article
8. Jiang W, Gupta RK, Deshpande MC, Schwendeman SP. Biodegradable poly(lactic-co-glycolic acid) microparticles for injectable delivery of vaccine antigens. Adv Drug Deliv Rev. 2005. 57:391–410.
Article
9. Johansen P, Men Y, Merkle HP, Gander B. Revisiting PLA/PLGA microspheres: an analysis of their potential in parenteral vaccination. Eur J Pharm Biopharm. 2000. 50:129–146.
Article
10. Karttunen J, Sanderson S, Shastri N. Detection of rare antigen-presenting cells by the lacZ T-cell activation assay suggests an expression cloning strategy for T-cell antigens. Proc Natl Acad Sci U S A. 1992. 89:6020–6024.
Article
11. Kocbek P, Obermajer N, Cegnar M, Kos J, Kristl J. Targeting cancer cells using PLGA nanoparticles surface modified with monoclonal antibody. J Control Release. 2007. 120:18–26.
Article
12. Lee JK, Lee MK, Yun YP, Kim Y, Kim JS, Kim YS, Kim K, Han SS, Lee CK. Acemannan purified from Aloe vera induces phenotypic and functional maturation of immature dendritic cells. Int Immunopharmacol. 2001. 1:1275–1284.
Article
13. Lee YH, Lee YR, Kim KH, Im SA, Song S, Lee MK, Kim Y, Hong JT, Kim K, Lee CK. Baccatin III, a synthetic precursor of taxol, enhances MHC-restricted antigen presentation in dendritic cells. Int Immunopharmacol. 2011. [Epub ahead of print].
Article
14. Lee YR, Yang IH, Lee YH, Im SA, Song S, Li H, Han K, Kim K, Eo SK, Lee CK. Cyclosporin A and tacrolimus, but not rapamycin, inhibit MHC-restricted antigen presentation pathways in dendritic cells. Blood. 2005. 105:3951–3955.
Article
15. Mundargi RC, Babu VR, Rangaswamy V, Patel P, Aminabhavi TM. Nano/micro technologies for delivering macromolecular therapeutics using poly(D,L-lactide-co-glycolide) and its derivatives. J Control Release. 2008. 125:193–209.
Article
16. Newman KD, Sosnowski DL, Kwon GS, Samuel J. Delivery of MUC1 mucin peptide by Poly(d,l-lactic-co-glycolic acid) microspheres induces type 1 T helper immune responses. J Pharm Sci. 1998. 87:1421–1427.
Article
17. Panyam J, Labhasetwar V. Biodegradable nanoparticles for drug and gene delivery to cells and tissue. Adv Drug Deliv Rev. 2003. 55:329–347.
Article
18. Sahoo SK, Panyam J, Prabha S, Labhasetwar V. Residual polyvinyl alcohol associated with poly (D,L-lactide-co-glycolide) nanoparticles affects their physical properties and cellular uptake. J Control Release. 2002. 82:105–114.
Article
19. Shen H, Ackerman AL, Cody V, Giodini A, Hinson ER, Cresswell P, Edelson RL, Saltzman WM, Hanlon DJ. Enhanced and prolonged cross-presentation following endosomal escape of exogenous antigens encapsulated in biodegradable nanoparticles. Immunology. 2006. 117:78–88.
Article
20. Sigal LJ, Crotty S, Andino R, Rock KL. Cytotoxic T-cell immunity to virus-infected non-haematopoietic cells requires presentation of exogenous antigen. Nature. 1999. 398:77–80.
Article
21. Venkataprasad N, Coombes AG, Singh M, Rohde M, Wilkinson K, Hudecz F, Davis SS, Vordermeier HM. Induction of cellular immunity to a mycobacterial antigen adsorbed on lamellar particles of lactide polymers. Vaccine. 1999. 17:1814–1819.
Article
22. Waeckerle-Men Y, Groettrup M. PLGA microspheres for improved antigen delivery to dendritic cells as cellular vaccines. Adv Drug Deliv Rev. 2005. 57:475–482.
Article
23. Yewdell JW, Haeryfar SM. Understanding presentation of viral antigens to CD8+ T cells in vivo: the key to rational vaccine design. Annu Rev Immunol. 2005. 23:651–682.
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