Original Article
Poly(lactic acid)–poly(ethylene glycol) nanoparticles provide sustained delivery of a Chlamydia trachomatis recombinant MOMP peptide and potentiate systemic adaptive immune responses in mice

https://doi.org/10.1016/j.nano.2014.02.009Get rights and content

Abstract

PLA–PEG [poly(lactic acid)–poly (ethylene glycol)], a biodegradable copolymer, is underexploited for vaccine delivery although it exhibits enhanced biocompatibility and slow release immune-potentiating properties. We document here successful encapsulation of M278, a Chlamydia trachomatis MOMP (major outer-membrane protein) peptide, within PLA–PEG nanoparticles by size (~ 73-100 nm), zeta potential (− 16 mV), smooth morphology, encapsulation efficiency (~ 60%), slow release pattern, and non-toxicity to macrophages. Immunization of mice with encapsulated M278 elicited higher M278-specific T-cell cytokines [Th1 (IFN-γ, IL-2), Th17 (IL-17)] and antibodies [Th1 (IgG2a), Th2 (IgG1, IgG2b)] compared to bare M278. Encapsulated-M278 mouse serum inhibited Chlamydia infectivity of macrophages, with a concomitant transcriptional down-regulation of MOMP, its cognate TLR2 and CD80 co-stimulatory molecule. Collectively, encapsulated M278 potentiated crucial adaptive immune responses, which are required by a vaccine candidate for protective immunity against Chlamydia. Our data highlight PLA–PEG's potential for vaccines, which resides in its slow release and potentiating effects to bolster immune responses.

From the Clinical Editor

This study highlights the potential of a PLA-PEG-based nanoparticle formulation containing a major outer membrane protein of chlamydia trachomatis in inducing a sustained enhanced immune response, paving the way to the development of a vaccination strategy against this infection.

Graphical Abstract

Nanoencapsulation of M278, a Chlamydia trachomatis recombinant MOMP (major outer-membrane protein) peptide within PLA–PEG copolymer (PLA–PEG–M278) provided its slow release that potentiated immune responses in BALB/c mice by sustained antigen presentation. Compared to bare M278, enhanced adaptive immune responses were triggered by encapsulated M278 in mice including specific T-cell cytokines and isotype antibodies, along with in vitro serum-mediated inhibition of Chlamydia infectivity of macrophages.

  1. Download : Download high-res image (183KB)
  2. Download : Download full-size image

Section snippets

Fabrication of nanoparticles

Recombinant MOMP-278 (M278) was cloned as previously reported15 and encapsulated in PLA–PEG nanoparticles using a modified water/oil/water double emulsion evaporation technique16, 17 and then lyophilized in the presence of 5% trehalose (as a stabilizer) to obtain encapsulated M278 (PLA–PEG–M278). Phosphate-buffered saline (PBS) was similarly encapsulated in PLA–PEG to serve as a negative control (PLA–PEG–PBS). All lyophilized nanoparticles were stored at − 80 °C in a sealed container until used.

Scanning electron microscopy (SEM) and transmission electron microscopy (TEM)

Physiochemical properties of nanoparticles

SEM and TEM microscopy techniques were employed to assess the morphology and size of nanoparticles. By SEM analysis, PLA–PEG–PBS appeared to be coagulated particles and in the conformation of what is referred to as PEGylated “brush” with rough outer surface23 (Figure 2, A). Magnification of this brush structure within PLA–PEG–M278 revealed a Web-like matrix that contained grape-like structures dispersed throughout (Figure 2, B). Additionally, TEM images of PLA–PEG–PBS (Figure 2, C) and

Discussion

Problems associated with conventional adjuvants for vaccines are protein denaturation and low bioavailability in vivo.25 PLA–PEG offers an attractive alternative to conventional adjuvants as a vaccine-delivery system because it can augment the immunogenicity of proteins by providing their controlled slow release to the immune system.1, 2, 26 Moreover, PLA–PEG can enhance the protein loading capacity, reduce its burst effect, prevent degradation, and increase the circulation time and

Acknowledgments

Special thanks go to Yvonne Williams, Lashaundria Lucas, Juwana Smith Henderson and Maiya Moore for their excellent administrative assistance; Eva Dennis for the graphical abstract, and Michael Miller, Auburn University, for assistance with SEM and TEM imaging.

References (50)

  • I. Gutierro et al.

    Size dependent immune response after subcutaneous, oral and intranasal administration of BSA loaded nanospheres

    Vaccine

    (2002)
  • A. Vila et al.

    Design of biodegradable particles for protein delivery

    J Control Release

    (2002)
  • A. Vonarbourg et al.

    Parameters influencing the stealthiness of colloidal drug delivery systems

    Biomaterials

    (2006)
  • M.J. Alonso et al.

    Biodegradable microspheres as controlled-release tetanus toxoid delivery systems

    Vaccine

    (1994)
  • R.M. Mainardes et al.

    Intranasal delivery of zidovudine by PLA and PLA–PEG blend nanoparticles

    Int J Pharm

    (2010)
  • A.J. Olive et al.

    CXCR3 and CCR5 are both required for T-cell-mediated protection against Chlamydia trachomatis infection in the murine genital mucosa

    Mucosal Immunol

    (2011)
  • S. Pal et al.

    Monoclonal immunoglobulin A antibody to the major outer membrane protein of the Chlamydia trachomatis mouse pneumonitis biovar protects mice against a chlamydial genital challenge

    Vaccine

    (1997)
  • J. De Souza Rebouças et al.

    Nanoparticulate adjuvants and delivery systems for allergen immunotherapy

    J Biomed Biotechnol

    (2012)
  • S. Essa et al.

    Improved antifungal activity of itraconazole-loaded PEG/PLA nanoparticles

    J Microencapsul

    (2013)
  • J. Sheringham et al.

    It matters what you measure: a systematic literature review examining whether young people in poorer socioeconomic circumstances are more at risk of chlamydia

    Sex Transm Infect

    (2013)
  • C.I. Champion et al.

    A vault nanoparticle vaccine induces protective mucosal immunity

    PLoS One

    (2009)
  • M. Sandhya et al.

    Nanoparticulate systems and other novel approaches in the diagnosis, prevention and treatment of HIV/AIDS

    Int J Pharm Sci

    (2010)
  • M.A. Taha et al.

    A peptide containing T-cell epitopes of Chlamydia trachomatis recombinant MOMP induces systemic and mucosal antibody responses in mice

    WJV

    (2011)
  • M.A. Taha et al.

    Biodegradable PLGA 85/15 nanoparticles as a delivery vehicle for Chlamydia trachomatis recombinant MOMP-187 peptide

    Nanotechnology

    (2012)
  • S.J. Fairley et al.

    Chlamydia trachomatis recombinant MOMP encapsulated in PLGA nanoparticles triggers primarily T helper-1 cellular and antibody immune responses in mice: a desirable candidate nanovaccine

    Int J Nanomedicine

    (2013)
  • Cited by (0)

    All authors declare no conflict of interest related to the submitted manuscript.

    Financial support: This research was supported by National Science Foundation-CREST (HRD-1241701), NSF-HBCU-UP (HRD-1135863) and National Institutes of Health-MBRS-RISE (1R25GM106995-01) grants.

    An abstract of this manuscript was presented at the TechConnect World Conference, May 11-16, 2013, Washington, DC, and at the 113th American Society for Microbiology General Meeting, May 18-21, 2013, Denver, CO.

    View full text