The characteristics, cellular uptake and intracellular trafficking of nanoparticles made of hydrophobically-modified chitosan

https://doi.org/10.1016/j.jconrel.2010.05.023Get rights and content

Abstract

It has been reported that nanoparticles (NPs) prepared by hydrophobically-modified polymers could accumulate passively in the tumor tissue; however, their cellular uptake mechanism and intercellular trafficking pathway have never been understood. This study was designed to address these concerns, using NPs prepared by a hydrophobically-modified chitosan (N-palmitoyl chitosan, NPCS). Molecular dynamic simulations found that a degree of substitution (DS) of 5% of palmitoyl groups on its backbone was sufficient to allow NPCS to form NPs, due to a significant increase in the intra- and intermolecular hydrophobic interactions. With an increase of DS, there were more palmitoyl groups present on the surface of NPs which were then able to interact with the cell membranes. A greater extent of cellular uptake of NPCS NPs was observed with increasing the DS on NPCS. The internalization of NPCS NPs was clearly related with the lipid raft-mediated routes; with increasing the DS on NPCS, the caveolae-mediated endocytosis became more important. The results obtained in the intracellular trafficking study showed that NPCS NPs entered cells via caveolae and transiently localized to caveosomes before trafficking to the endosomal pathway. These results suggest that the prepared NCPS NPs may serve as a carrier for intracellular delivery of therapeutic agents.

Introduction

Chitosan (CS), a natural-origin polysaccharide, is biodegradable, non-toxic and soft-tissue compatible, and thus has been used extensively in biomedical applications [1]. It is known that the pKa of CS is approximately 6.5 [1], [2]. In aqueous media at pH 7.4, CS was found to form dissociated precipitates due to deprotonation of its amine groups. To enhance its intermolecular contact, we conjugated a hydrophobic palmitoyl group onto the free amine groups of CS in a previous study [3]. The synthesized N-palmitoyl CS (NPCS) in an aqueous environment was demonstrated to exhibit a rapid nanostructure transformation within a narrow pH range through a proper balance between charge repulsion and hydrophobic interaction. Subcutaneous injection of aqueous NPCS into a rat model resulted in rapid formation of a massive hydrogel at the location of injection.

In dilute aqueous media, we found that NPCS polymers were able to self-assemble into nanoparticles (NPs), due to the hydrophobic interaction between their conjugated palmitoyl groups. Hydrophobically-modified polymers have been used to fabricate NPs as a drug-delivery vehicle for therapeutic applications [4], [5], [6]. Previous studies have shown that these NPs could accumulate passively in the tumor tissue [4], [5]. The cellular uptake mechanism and intracellular fate of 5β-cholanic-acid conjugated glycol CS, a hydrophobic glycol CS (HGC), has been reported by Nam et al. [7]; interestingly, several distinct uptake pathways were identified to be involved in the internalization of HGC with a single degree of substitution (DS). Whether different DS (or hydrophobicity) could affect the endocytosis of hydrophobically-modified polymers remains to be understood. In the present study, we prepared NPs of NPCS with different DS; their cellular uptake and the potential endocytosis and intracellular trafficking pathways were investigated herein.

A few synthetic methods have been developed to conjugate hydrophobic side chains on the CS backbone. Nevertheless, these synthetic methods led to either a relatively low DS of 1–5% [8], [9], [10], [11], [12] or caused an uncontrollable DS and produced undesired by-products [13]. In this study, we employed a method which was able to produce NPCS with relatively high and controllable DS (up to 20% DS). The synthesis was accomplished in a single-step reaction which had been used in forming amide bonds in the synthesis of peptides [3], [14].

The NPs prepared by the synthesized NPCS polymers with different DS were characterized using dynamic light scattering (DLS) and molecular dynamic (MD) simulations. The cytotoxicity of NPs was evaluated by the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay as well as the lactate dehydrogenase (LDH) assay. The cellular internalization efficiency, potential endocytosis mechanism and intracellular trafficking pathway of test NPs were investigated via a flow cytometer and a confocal laser scanning microscope (CLSM).

Section snippets

Materials

CS (viscosity 5 mPa s, 0.5% in 0.5% acetic acid at 20 °C, MW 50 kDa) with a degree of deacetylation of approximately 85% was purchased from Koyo Chemical Co. Ltd. (Tokyo, Japan). Palmitic acid N-hydroxysuccinimide ester was obtained from Sigma-Aldrich (St. Louis, MO, USA). N-hydroxysuccinimide (NHS)-functionalized cyanine 5 (Cy5-NHS) and fluorescein (fluorescein-NHS) were acquired from Amersham Biosciences (Piscataway, NJ, USA) and Thermo Scientific (Chicago, IL, USA), respectively. All other

Results and discussion

Hydrophobically-modified polymers, which are water-soluble polymers bearing hydrophobic side chains, have found an increasing number of biological applications [30], [31], [32]. Because the contact between the hydrophobic side chains and water is energetically unfavorable, the hydrophobically-modified polymers have a strong tendency to self-assemble into aggregates on the nanometer scale in an aqueous environment [33]. In this study, we synthesized hydrophobically-modified CS (NPCS) with

Conclusions

In conclusion, the extent of cellular uptake of NPs was significantly enhanced with increasing the DS on NPCS. The internalization of NPCS NPs was clearly related with the lipid raft-mediated routes. With increasing the DS on NPCS polymers, the caveolae-mediated endocytosis became more important. The internalized NPs transiently associate with CAV1 at cell membranes and at a peripheral CAV1-positive structure coupled with caveosomes before trafficking to the endosomal pathway.

Acknowledgements

This work was supported by a grant from the National Science Council (NSC 98-2120-M-007-007), Taiwan, Republic of China.

References (47)

  • N. Weber et al.

    Characterization of carbosilane dendrimers as effective carriers of siRNA to HIV-infected lymphocytes

    J. Control. Release

    (2008)
  • Y.P. Ho et al.

    Evaluating the intracellular stability and unpacking of DNA nanocomplexes by quantum dots-FRET

    J. Control. Release

    (2006)
  • O.P. Perumal et al.

    The effect of surface functionality on cellular trafficking of dendrimers

    Biomaterials

    (2008)
  • Y. Choi et al.

    Synthesis and functional evaluation of DNA-assembled polyamidoamine dendrimer clusters for cancer cell-specific targeting

    Chem. Biol.

    (2005)
  • J. Kötz et al.

    Self-assembled polyelectrolyte systems

    Prog. Polym. Sci.

    (2001)
  • P.J. Bond et al.

    Coarse-grained molecular dynamics simulations of membrane proteins and peptides

    J. Struct. Biol.

    (2007)
  • S.K. Lai et al.

    Privileged delivery of polymer nanoparticles to the perinuclear region of live cells via a non-clathrin, non-degradative pathway

    Biomaterials

    (2007)
  • F.P. Seib et al.

    Comparison of the endocytic properties of linear and branched PEIs, and cationic PAMAM dendrimers in B16f10 melanoma cells

    J. Control. Release

    (2007)
  • S.K. Sahoo et al.

    Residual polyvinyl alcohol associated with poly (D, L-lactide-co-glycolide) nanoparticles affects their physical properties and cellular uptake

    J. Control. Release

    (2002)
  • S. Arni et al.

    Association of GAP-43 with detergent-resistant membranes requires two palmitoylated cysteine residues

    J. Biol. Chem.

    (1998)
  • L.M. Bareford et al.

    Endocytic mechanisms for targeted drug delivery

    Adv. Drug Deliv. Rev.

    (2007)
  • M.N.V.R. Kumar et al.

    Chitosan chemistry and pharmaceutical perspectives

    Chem. Rev.

    (2004)
  • J.H. Park et al.

    Self-assembled nanoparticles based on glycol chitosan bearing hydrophobic moieties as carriers for doxorubicin: in vivo biodistribution and anti-tumor activity

    Biomaterials

    (2006)
  • Cited by (194)

    View all citing articles on Scopus
    View full text