Elsevier

Carbohydrate Polymers

Volume 223, 1 November 2019, 115128
Carbohydrate Polymers

Polyethylenimine-coated PLGA nanoparticles-encapsulated Angelica sinensis polysaccharide as an adjuvant to enhance immune responses

https://doi.org/10.1016/j.carbpol.2019.115128Get rights and content

Highlights

  • ASP-PLGA-PEI nanoparticles has a good stability over 28 days.

  • ASP-PLGA-PEI showed excellent performance in the activation of macrophages.

  • ASP-PLGA-PEI effectively adsorbed antigen and promote antigen uptake by macrophages.

  • ASP-PLGA-PEI-PCV2 antigen significantly induced PCV2-specific IgG immune response.

  • ASP-PLGA-PEI-PCV2 elicit a mixed Th1/Th2 response with Th1 bias compared with other groups.

Abstract

Nanoparticle delivery systems have been widely investigated as new vaccines strategy to enhance the immune responses to antigens against infectious diseases. The positively charged nanoparticles could efficiently improve the immune responses due to targeting and activating the antigen-presenting cells. In this study, the immunopotentiator Angelica sinensis polysaccharide (ASP) was encapsulated into Poly (lactic-co-glycolic acid) (PLGA) nanoparticles, and the polyethylenimine, one of the cationic polymers, was used to coat nanoparticles to develop a new nanoparticle delivery system (ASP-PLGA-PEI) with positively charged. The ASP-PLGA-PEI nanoparticles significantly activated macrophages, and promoted the expression of the MHCII and CD86 and the production of IL-1β and IL-12p70 cytokines of macrophages. Furthermore, the antigen adsorbed on the surface of the ASP-PLGA-PEI nanoparticles enhanced the antigen uptake by macrophages. Moreover, the mice immunized with PCV2 antigen adsorbed ASP-PLGA-PEI nanoparticles significantly enhanced PCV2-specific IgG immune response and the levels of cytokines, induced a mixed Th1/Th2 immune response with Th1 bias compared with other groups. These findings demonstrate that the positively charged nanoparticles (ASP-PLGA-PEI) have the potential to serve as an effective vaccine delivery and adjuvant system to induce vigorous and long-term immune responses.

Introduction

Vaccination plays a crucial role in preventing infectious and viral diseases because it could induce a vigorous antigen-specific immune response for long-term protection and therapy (Liu, Cao et al., 2016, 2016b; Sarti et al., 2011). Compared to traditional vaccines, the protein subunit vaccines are considered to be safer but with poor immunogenicity. The adjuvants are required to augment resultant humoral and cell-mediated immune responses (Liu, Cao et al., 2016; Zhang, Wang et al., 2014). Aluminum, as a conventional and commercial adjuvant, had used in vaccines for many years. However, the Aluminum has some disadvantages, including local and systemic side effects, poorly cell-mediated immune responses and inefficiency for some antigens (Chen et al., 2014; Courant et al., 2017; Kool et al., 2008). Therefore, it is necessary to design safer and more effective adjuvants to induce strong humoral immune responses and stimulate cell-mediated immune responses.

Angelica sinensis (AS), a Chinese herbal medicine, has been traditionally used to treat anemia, constipation, hepatic fibrosis, cardiovascular disease and gynecological diseases for thousands of years (Hook, 2014; Hua, Xue, Zhang, Wei, & Ji, 2014). Angelica sinensis polysaccharide (ASP), one of the main active components, possess varieties of pharmacological activities, such as hematopoietic effect, hepatoprotective effect, antioxidant potency, antitumor activity, radioprotective effect, and immunologic enhancement (Ji et al., 2014; Lee, Hsieh, Chen, & Chiang, 2012; Zhang et al., 2016; Zhao et al., 2012; Zhuang et al., 2016). It has been reported that the ASP was composed of fucose, galactose, glucose, arabinose, rhamnose, and xylose and in a molar ratio of 1.0:13.6:15.0:8.7:21.3:3.7 (Wang, Ding, Zhu, He, & Fang, 2003), and the neutral ASP was mainly composed of galactose, arabinose and glucose (Zhao et al., 2012), in consistent with the results reported by others (Sun, Tang, Gu, & Li, 2005; Zhang, Cheng, Wang, Zhang, & Wang, 2014). It was found that ASP had a backbone composed of 1,4-α-D-glucopyranosyl residues, with branches attached to O-6 of some residues. The branches were composed of 1,6-α-D-Glcp residues, and terminated with β-L-arabinofuranose residues (Cao, Li, Liu, Yang et al., 2006, 2006b). Recently, ASP has been widely used as immunostimulant and adjuvant to improve the immune activity (Wang, Ge, Li, Guan, & Li, 2016; Yang, Jia, Meng, Wu, & Mei, 2006). However, ASP has some disadvantages such as non-focused action scope, fast-metabolism and low bioavailability, which limit its application.

In recent years, the nanoparticles adjuvant delivery systems have drawn growing attention because the nanoparticles could protect the antigen and drugs from degradation, increase drug bioavailability, facilitate antigen uptake by antigen presenting cells (APCs) and enhance immune responses (Canadas et al., 2016; Divya et al., 2018; Du et al., 2017; Vijayakumar & Vaseeharan, 2018; Vijayakumar et al., 2017; Yue & Ma, 2015). Nanoparticles delivery systems also have the potential to regulate the antigen presentation pathway (Yue & Ma, 2015; Zupancic et al., 2017). Several studies have proved that nanoparticles with positive surface charge could increase the adsorption of antigen, enhance the interaction with negatively charged cell membrane and facilitate the intracellular uptake of antigen by APCs such as dendritic cells (DCs) and macrophages to modulate the immune responses (Chen et al., 2014; Liu, Ma et al., 2016; Salvador et al., 2015; Zupancic et al., 2017). Poly (lactic-co-glycolic acid) (PLGA), a biodegradable and biocompatible polymer, has been extensively used to prepare nanoparticles for drug and vaccine delivery with high safety and controlled release (Hafner, Corthesy, Textor, & Merkle, 2016; Salvador et al., 2015). Numerous researches have demonstrated that PLGA nanoparticles delivery systems could protect drug and antigen, provide a controlled and persistent release of drug and antigen and co-deliver drug and antigen to the same APCs (Danhier et al., 2012; Sarti et al., 2011; Wang, Tan, Keegan, Barry, & Heffernan, 2014). However, the PLGA nanoparticles with no modification are often negatively charged, which limits the adjuvant activity of the nanoparticles. Cationic polymers had been widely used as transfection agents and could activate the macrophages and DCs to induce immune responses (Mulens-Arias, Rojas, Perez-Yague, Morales, & Barber, 2015). The polyethylenimine (PEI), one of the widely used cationic polymers, could modify the surface charge of the PLGA nanoparticles (Salvador et al., 2015). It has been proved that the PEI-coated nanoparticles, used as the drug delivery systems, could promote the cellular uptake efficiency and improve immune responses (Chen et al., 2014; Salvador et al., 2015; Yu et al., 2016).

In our previous researches, the ASP was encapsulated into PLGA nanoparticles to develop a drug delivery system (ASP-PLGA nanoparticles) to improve the immunological enhancement of ASP (Gu et al., 2018). In addition, it has been reported the OVA encapsulated into ASP-PLGA nanoparticles as a vaccine delivery system could stimulate strong humoral responses and cellular immune responses (Gu et al., 2019). However, the ASP-PLGA nanoparticles had shown poorly effects on the activation of APCs in our preliminary experiments. In this study, we had developed a stable and well-characterized cationic nanoparticle delivery system (ASP-PLGA-PEI), and we hypothesized that ASP-PLGA-PEI nanoparticles would function as an effective adjuvant and vaccine system to target and activate the APCs, and induce strong humoral and cell-mediated immune responses. This hypothesis was tested by investigating the effects of ASP-PLGA-PEI nanoparticles on activation of macrophages and antigen uptake by macrophages. Furthermore, we utilized the PCV2 antigen adsorbed on ASP-PLGA-PEI nanoparticles to evaluate the ability to induce humoral and cell-mediated immune responses.

Section snippets

Materials

ASP (95%, CY170618) was obtained from Shanxi Ciyuan Biotechnology Co., Ltd (Shanxi, China). PLGA (75:25, MW 18 kDa) was purchased from Jinan Daigang Biomaterial Co., Ltd (Shandong, China). Pluronic F68 (F68) and Sephadex G-50 were obtained from Shanghai Yuanye Biotechnology Co., Ltd (Shanghai, China). PEI (MW 25 kDa) was purchased from Sigma-Aldrich (USA). Anti-Mouse-MHCII-FITC anti-Mouse-CD86-PE antibodies were purchased from eBioscience Inc. (Thermo Fisher Scientific, USA). OVA-FITC and the

FT-IR spectra of ASP

The FT-IR spectra of the ASP were shown in Fig. 1. The band at around 3400 cm−1 was due to the stretching vibration of Osingle bondH. The weak peak at around 2932 cm−1 was due to the asymmetrical Csingle bondH stretching and bending vibration. The band at around 1632 cm-1 was attributed to the bound water (Zhang et al., 2016). The absorption peaks at around 1416 cm−1 and 1055 cm-1 were characteristic bands of the carboxylic group. The peaks at 1400–1200 cm-1 were also the presence of the Csingle bondH bonds. The wave number

Discussion

Numerous studies have shown that Angelica sinensis polysaccharide (ASP) possess the immunomodulatory activity related with T-lymphocyte activation (Jin, Zhao, Huang, Xu, & Shang, 2012; Sun et al., 2005; Yang, Jia et al., 2006). The ASP was biologically safe and had been widely used as immunopotentiator and adjuvant to enhance the immune responses (Wang et al., 2016). Recently, the vaccine delivery and adjuvant systems based PLGA nanoparticles have been intensively investigated due to their

Conclusions

Herein, we successfully engineered a simple, efficient well-characterized and good stability nanoparticles delivery system with positive surface charge. The ASP-PLGA-PEI nanoparticles showed excellent performance in the activation of macrophages. The results indicated that ASP-PLGA-PEI nanoparticles could be a useful strategy for effective antigen adsorption and efficiently enhance the antigen uptake by macrophages. To further investigate the adjuvant activity of the ASP-PLGA-PEI nanoparticles,

Acknowledgments

The project was supported by the National Natural Science Foundation of China (Grant No. 31872509, 31672596), the Fundamental Research Funds for the Central Universities (Grant No. KYZ201844) and A Project Funded by the Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD). We are grateful to all other staff in the Institute of Traditional Chinese Veterinary Medicine of Nanjing Agricultural University for their assistance in this study.

References (54)

  • P. Gu et al.

    Optimization of angelica sinensis Polysaccharide-loaded Poly (lactic-co-glycolicacid) nanoparticles by RSM and its immunological activity in vitro

    International Journal of Biological Macromolecules

    (2018)
  • N. Gunday Tureli et al.

    Optimization of ciprofloxacin complex loaded PLGA nanoparticles for pulmonary treatment of cystic fibrosis infections: Design of experiments approach

    International Journal of Pharmaceutics

    (2016)
  • A.M. Hafner et al.

    Surface-assembled poly(I:C) on PEGylated PLGA microspheres as vaccine adjuvant: APC activation and bystander cell stimulation

    International Journal of Pharmaceutics

    (2016)
  • I.L. Hook

    Danggui to Angelica sinensis root: Are potential benefits to European women lost in translation? A review

    Journal of Ethnopharmacology

    (2014)
  • Y. Hua et al.

    Metabonomics study on the hepatoprotective effect of polysaccharides from different preparations of Angelica sinensis

    Journal of Ethnopharmacology

    (2014)
  • P. Ji et al.

    Metabolomics research on the hepatoprotective effect of Angelica sinensis polysaccharides through gas chromatography-mass spectrometry

    Journal of Chromatography. B, Analytical Technologies in the Biomedical and Life Sciences

    (2014)
  • M. Jin et al.

    Isolation, structure and bioactivities of the polysaccharides from Angelica sinensis (Oliv.) Diels: A review

    Carbohydrate Polymers

    (2012)
  • S.Y. Kim et al.

    Synthetic vaccine nanoparticles target to lymph node triggering enhanced innate and adaptive antitumor immunity

    Biomaterials

    (2017)
  • N.K. Kunda et al.

    Pulmonary dry powder vaccine of pneumococcal antigen loaded nanoparticles

    International Journal of Pharmaceutics

    (2015)
  • J.G. Lee et al.

    Hematopoietic and myeloprotective activities of an acidic Angelica sinensis polysaccharide on human CD34+ stem cells

    Journal of Ethnopharmacology

    (2012)
  • L. Liu et al.

    Immune responses to vaccines delivered by encapsulation into and/or adsorption onto cationic lipid-PLGA hybrid nanoparticles

    Journal of Controlled Release

    (2016)
  • Y. Liu et al.

    The effect of Gd@C82(OH)22 nanoparticles on the release of Th1/Th2 cytokines and induction of TNF-alpha mediated cellular immunity

    Biomaterials

    (2009)
  • Z. Liu et al.

    Activation effect of Ganoderma lucidum polysaccharides liposomes on murine peritoneal macrophages

    International Journal of Biological Macromolecules

    (2016)
  • V. Mulens-Arias et al.

    Polyethylenimine-coated SPIONs trigger macrophage activation through TLR-4 signaling and ROS production and modulate podosome dynamics

    Biomaterials

    (2015)
  • A. Salvador et al.

    Design and evaluation of surface and adjuvant modified PLGA microspheres for uptake by dendritic cells to improve vaccine responses

    International Journal of Pharmaceutics

    (2015)
  • F. Sarti et al.

    In vivo evidence of oral vaccination with PLGA nanoparticles containing the immunostimulant monophosphoryl lipid A

    Biomaterials

    (2011)
  • A.L. Siefert et al.

    Artificial bacterial biomimetic nanoparticles synergize pathogen-associated molecular patterns for vaccine efficacy

    Biomaterials

    (2016)
  • Cited by (82)

    View all citing articles on Scopus
    View full text