Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Pharm ; 20(2): 953-970, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36583936

ABSTRACT

Mucosal surfaces of the lungs represent a major site of entry for airborne pathogens, and pulmonary administration of vaccines is an attractive strategy to induce protective mucosal immunity in the airways. Recently, we demonstrated the potential of pulmonary vaccination with the tuberculosis subunit antigen H56 adjuvanted with the cationic liposomal adjuvant formulation CAF01, which consists of the cationic lipid dimethyldioctadecylammonium (DDA) bromide and the synthetic cord factor trehalose-6,6'-dibehenate. However, the cationic charge of DDA represents a major safety challenge. Hence, replacing DDA with a safer zwitterionic or anionic phospholipid is an attractive approach to improve vaccine safety, but the effect of liposomal surface charge on the induction of mucosal immunity after airway immunization is poorly understood. Here, we investigated the effect of surface charge by replacing the cationic DDA component of CAF01 with zwitterionic dipalmitoylphosphatidylcholine (DPPC) or anionic dipalmitoylphosphatidylglycerol (DPPG), and we show that charge modification enhances antigen-specific pulmonary T-cell responses against co-formulated H56. We systematically replaced DDA with either DPPC or DPPG and found that these modifications resulted in colloidally stable liposomes that have similar size and morphology to unmodified CAF01. DPPC- or DPPG-modified CAF01 displayed surface charge-dependent protein adsorption and induced slightly higher follicular helper T cells and germinal center B cells in the lung-draining lymph nodes than unmodified CAF01. In addition, modified CAF01 induced significantly higher levels of H56-specific Th17 cells and polyfunctional CD4+ T cells in the lungs, as compared to unmodified CAF01. However, the strong H56-specific humoral responses induced by CAF01 in the lungs and spleen were not influenced by surface charge. Hence, these results provide insights into the importance of surface charge for liposomal adjuvant function and can also guide the design of safe pulmonary subunit vaccines against other mucosal pathogens.


Subject(s)
Adjuvants, Immunologic , Liposomes , Animals , Mice , Immunization , Vaccination , Vaccines, Subunit , Adjuvants, Pharmaceutic , Mice, Inbred C57BL , Quaternary Ammonium Compounds
2.
Int J Mol Sci ; 22(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638944

ABSTRACT

Natural polysaccharides have shown promising effects on the regulation of immunity in animals. In this study, we examined the immune stimulatory effect of intranasally administered Codium fragile polysaccharides (CFPs) in mice. Intranasal administration of CFPs in C57BL/6 mice induced the upregulation of surface activation marker expression in macrophages and dendritic cells (DCs) in the mediastinal lymph node (mLN) and the production of interleukin-6 (IL-6), IL-12p70, and tumor necrosis factor-α in bronchoalveolar lavage fluid. Moreover, the number of conventional DCs (cDCs) was increased in the mLNs by the upregulation of C-C motif chemokine receptor 7 expression, and subsets of cDCs were also activated following the intranasal administration of CFP. In addition, the intranasal administration of CFPs promoted the activation of natural killer (NK) and T cells in the mLNs, which produce pro-inflammatory cytokines and cytotoxic mediators. Finally, daily administration of CFPs inhibited the infiltration of Lewis lung carcinoma cells into the lungs, and the preventive effect of CFPs on tumor growth required NK and CD8 T cells. Furthermore, CFPs combined with anti-programmed cell death-ligand 1 (PD-L1) antibody (Ab) improved the therapeutic effect of anti-PD-L1 Ab against lung cancer. Therefore, these data demonstrated that the intranasal administration of CFP induced mucosal immunity against lung cancer.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antineoplastic Agents/administration & dosage , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/therapy , Chlorophyta/chemistry , Immunity, Mucosal , Immunotherapy/methods , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Phytotherapy/methods , Plant Extracts/administration & dosage , Polysaccharides/administration & dosage , Administration, Intranasal/methods , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Dendritic Cells/immunology , Disease Models, Animal , Female , Killer Cells, Natural/immunology , Lung Neoplasms/pathology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL
3.
Int J Mol Sci ; 22(17)2021 Aug 24.
Article in English | MEDLINE | ID: mdl-34502035

ABSTRACT

Although fucoidan, a well-studied seaweed-extracted polysaccharide, has shown immune stimulatory effects that elicit anticancer immunity, mucosal adjuvant effects via intranasal administration have not been studied. In this study, the effect of Ecklonia cava-extracted fucoidan (ECF) on the induction of anti-cancer immunity in the lung was examined by intranasal administration. In C57BL/6 and BALB/c mice, intranasal administration of ECF promoted the activation of dendritic cells (DCs), natural killer (NK) cells, and T cells in the mediastinal lymph node (mLN). The ECF-induced NK and T cell activation was mediated by DCs. In addition, intranasal injection with ECF enhanced the anti-PD-L1 antibody-mediated anti-cancer activities against B16 melanoma and CT-26 carcinoma tumor growth in the lungs, which were required cytotoxic T lymphocytes and NK cells. Thus, these data demonstrated that ECF functioned as a mucosal adjuvant that enhanced the immunotherapeutic effect of immune checkpoint inhibitors against metastatic lung cancer.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Laminaria/chemistry , Lung Neoplasms/drug therapy , Polysaccharides/therapeutic use , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Administration, Intranasal , Animals , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/immunology , Drug Combinations , Female , Immune Checkpoint Inhibitors/administration & dosage , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lung Neoplasms/pathology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Metastasis , Plant Extracts , Polysaccharides/administration & dosage , Polysaccharides/pharmacology
4.
Microb Cell Fact ; 19(1): 42, 2020 Feb 19.
Article in English | MEDLINE | ID: mdl-32075660

ABSTRACT

BACKGROUND: Spore-forming bacteria of the Bacillus genus are widely used probiotics known to exert their beneficial effects also through the stimulation of the host immune response. The oral delivery of B. toyonensis spores has been shown to improve the immune response to a parenterally administered viral antigen in mice, suggesting that probiotics may increase the efficiency of systemic vaccines. We used the C fragment of the tetanus toxin (TTFC) as a model antigen to evaluate whether a treatment with B. toyonensis spores affected the immune response to a mucosal antigen. RESULTS: Purified TTFC was given to mice by the nasal route either as a free protein or adsorbed to B. subtilis spores, a mucosal vaccine delivery system proved effective with several antigens, including TTFC. Spore adsorption was extremely efficient and TTFC was shown to be exposed on the spore surface. Spore-adsorbed TTFC was more efficient than the free antigen in inducing an immune response and the probiotic treatment improved the response, increasing the production of TTFC-specific secretory immunoglobin A (sIgA) and causing a faster production of serum IgG. The analysis of the induced cytokines indicated that also the cellular immune response was increased by the probiotic treatment. A 16S RNA-based analysis of the gut microbial composition did not show dramatic differences due to the probiotic treatment. However, the abundance of members of the Ruminiclostridium 6 genus was found to correlate with the increased immune response of animals immunized with the spore-adsorbed antigen and treated with the probiotic. CONCLUSION: Our results indicate that B. toyonensis spores significantly contribute to the humoral and cellular responses elicited by a mucosal immunization with spore-adsorbed TTFC, pointing to the probiotic treatment as an alternative to the use of adjuvants for mucosal vaccinations.


Subject(s)
Bacillus/immunology , Immunity, Mucosal , Probiotics/therapeutic use , Spores, Bacterial/immunology , Tetanus Toxin/administration & dosage , Administration, Intranasal , Animals , Bacillus subtilis/immunology , Immunization , Male , Mice , Mice, Inbred C57BL
5.
Appl Microbiol Biotechnol ; 103(15): 5947-5955, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31175431

ABSTRACT

Over the past two decades, lactic acid bacteria (LAB) have been intensively studied as potential bacterial carriers for therapeutic materials, such as vaccine antigens, to the mucosal tissues. LAB have several attractive advantages as carriers of mucosal vaccines, and the effectiveness of LAB vaccines has been demonstrated in numerous studies. Research on LAB vaccines to date has focused on whether antigen-specific immunity, particularly antibody responses, can be induced. However, with recent developments in immunology, microbiology, and vaccinology, more detailed analyses of the underlying mechanisms, especially, of the induction of cell-mediated immunity and memory cells, have been required for vaccine development and licensure. In this mini-review, we will discuss the issues, including (i) immune responses other than antibody production, (ii) persistence of LAB vaccine immunity, (iii) comparative evaluation of LAB vaccines with any existing or reference vaccines, (iv) strategies for increasing the effectiveness of LAB vaccines, and (iv) effects of microbiota on the efficacy of LAB vaccines. Although these issues have been rarely studied or discussed to date in relation to LAB vaccine research, further understanding of them is critical for the practical application of LAB vaccine systems.


Subject(s)
Drug Carriers , Immunity, Mucosal , Lactobacillales/genetics , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Administration, Mucosal , Biomedical Research/trends , Technology, Pharmaceutical/trends , Vaccines, Synthetic/administration & dosage
6.
Mol Pharm ; 15(9): 4226-4234, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30107747

ABSTRACT

Infections remain a major threat to human lives. To overcome the threat caused by pathogens, mucosal vaccines are considered a promising strategy. However, no inactivated and/or subunit mucosal vaccine has been approved for human use, largely because of the lack of a safe and effective mucosal adjuvant. Here, we show that enzymatically synthesized polymeric caffeic acid (pCA) can act as a potent mucosal adjuvant in mice. Intranasal administration of ovalbumin (OVA) in combination with pCA resulted in the induction of OVA-specific mucosal IgA and serum IgG, especially IgG1. Importantly, pCA was synthesized from caffeic acid and horseradish peroxidase from coffee beans and horseradish, respectively, which are commonly consumed. Therefore, pCA is believed to be a highly safe material. In fact, administration of pCA did not show distinct toxicity in mice. These data indicate that pCA has merit for use as a mucosal adjuvant for nasal vaccine formulations.


Subject(s)
Adjuvants, Immunologic/chemistry , Caffeic Acids/chemistry , Caffeic Acids/immunology , Animals , Armoracia/chemistry , Cell Migration Assays, Leukocyte , Coffee/chemistry , Enzyme-Linked Immunosorbent Assay , Female , Horseradish Peroxidase/metabolism , Immunoglobulin A/metabolism , Immunoglobulin G/blood , Lignin/metabolism , Liposomes/administration & dosage , Liposomes/chemistry , Mice , Mice, Inbred BALB C
7.
Chem Pharm Bull (Tokyo) ; 66(4): 375-381, 2018.
Article in English | MEDLINE | ID: mdl-29607903

ABSTRACT

We have been investigating the potential of oligoarginine-linked polymers as an adjuvant for mucosal vaccination that induces immunoglobulin G (IgG) in systemic circulation and immunoglobulin A (IgA) secreted on the mucosa. Our latest infection experiments demonstrated that mice immunized nasally with a mixture of inactivated influenza viruses and poly(N-vinylacetamide-co-acrylic acid) (PNVA-co-AA) modified with D-octaarginine were perfectly protected from homologous virus infection. On the contrary, virus infection was observed in mice immunized with the antigen alone. This difference was presumably due to insignificant induction of secreted IgA on the nasal mucosa in the latter mice. Since it was unclear whether the current induction level was sufficient for heterologous virus infection, we evaluated the effects of the chemical structures of oligoarginines conjugated to PNVA-co-AA on induction of intranasal IgA. The number and optical activity of the arginine residues and the degree of modification with oligoarginines in the polymer backbone were listed as a factor that would influence IgA induction. Mouse experiments revealed that maximization of the modification resulted in an increase in adjuvant activities of oligoarginine-linked polymers most effectively. Glycine segments inserted between oligoarginines and the polymer backbone were a prerequisite for the maximization. The highest IgA level was observed when antigens were coadministered with diglycine-D-octaarginine-linked PNVA-co-AA.


Subject(s)
Adjuvants, Immunologic/chemistry , Antibodies/immunology , Arginine/chemistry , Biocompatible Materials/chemistry , Mucous Membrane/immunology , Nasal Cavity/immunology , Polymers/chemistry , Animals , Antibodies/chemistry , Arginine/analogs & derivatives , Female , Mice , Mice, Inbred BALB C , Molecular Structure , Mucous Membrane/chemistry
8.
Virol J ; 14(1): 44, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28253901

ABSTRACT

Despite availability of annual influenza vaccines, influenza causes significant morbidity and mortality in the elderly. This is at least in part a result of immunosenescence; the age-dependent decrease in immunological competence that results in greater susceptibility to infections and reduced responses to vaccination. To improve protective immune responses in this age group, new vaccines strategies, such as the use of adjuvants, are needed. Here, we evaluated the mucosal vaccine adjuvant Endocine™, formulated with split influenza antigen and administered intranasally in aged (20-month old) mice. Humoral immune responses were assessed and compared to unadjuvanted intranasal and subcutaneous vaccines. We show that formulation with Endocine™ significantly enhances hemagglutination inhibition (HI) titers, as well as serum IgG and mucosal IgA antibody titers, compared to both types of unadjuvanted vaccines. Thus, our results indicate that intranasal vaccination with Endocine™ is a possible approach for the development of mucosal influenza vaccines for the elderly.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antibody Formation , Antigens, Viral/administration & dosage , Antigens, Viral/immunology , Immunity, Mucosal , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Administration, Intranasal , Animals , Antibodies, Viral/blood , Female , Hemagglutination Tests , Immunoglobulin A/analysis , Immunoglobulin G/blood , Mice, Inbred BALB C , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
9.
Mol Pharm ; 14(5): 1469-1481, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28319404

ABSTRACT

The diameter of biodegradable particles used to coencapsulate immunostimulants and subunit vaccines affects the magnitude of memory CD8+ T cells generated by systemic immunization. Possible effects on the magnitude of CD8+ T cells generated by mucosal immunization or memory subsets that potentially correlate more strongly with protection against certain pathogens, however, are unknown. In this study, we conjugated our novel host-derived mucosal immunostimulant, EP67, to the protective MCMV CTL epitope, pp89, through a lysosomal protease-labile double arginine linker (pp89-RR-EP67) and encapsulated in PLGA 50:50 micro- or nanoparticles. We then compared total magnitude, effector/central memory (CD127/KRLG1/CD62L), and IFN-γ/TNF-α/IL-2 secreting subsets of pp89-specific CD8+ T cells as well as protection of naive female BALB/c mice against primary respiratory infection with MCMV 21 days after respiratory immunization. We found that decreasing the diameter of encapsulating particle from ∼5.4 µm to ∼350 nm (i) increased the magnitude of pp89-specific CD8+ T cells in the lungs and spleen; (ii) partially changed CD127/KLRG1 effector memory subsets in the lungs but not the spleen; (iii) changed CD127/KRLG1/CD62L effector/central memory subsets in the spleen; (iv) changed pp89-responsive IFN-γ/TNF-α/IL-2 secreting subsets in the lungs and spleen; (v) did not affect the extent to which encapsulation increased efficacy against primary MCMV respiratory infection over unencapsulated pp89-RR-EP67. Thus, although not observed under our current experimental conditions with MCMV, varying the diameter of nanoscale biodegradable particles may increase the efficacy of mucosal immunization with coencapsulated immunostimulant/subunit vaccines against certain pathogens by selectively increasing memory subset(s) of CD8+ T cells that correlate the strongest with protection.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Nanoparticles/chemistry , Nanospheres/chemistry , Vaccines, Subunit/chemistry , Animals , Cytomegalovirus/immunology , Female , Immunity, Mucosal/immunology , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Tumor Necrosis Factor-alpha/metabolism , Vaccines, Subunit/immunology
10.
Bioorg Med Chem ; 25(6): 1747-1755, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28237555

ABSTRACT

A series of new simplified oleanolic acid saponins with a glycosyl ester moiety at C28, were efficiently prepared. Furthermore, the effect of nasal administration of the synthetic oleanolic acid saponins on the nasal anti-influenza virus antibody titer against secondary nasal inoculation of the influenza split vaccine was examined. The result revealed cinnamoyl saponin as a suitable candidate vaccine adjuvant.


Subject(s)
Adjuvants, Immunologic/chemical synthesis , Adjuvants, Immunologic/pharmacology , Influenza Vaccines/administration & dosage , Nasal Mucosa/drug effects , Oleanolic Acid/chemistry , Saponins/chemical synthesis , Saponins/pharmacology , Adjuvants, Immunologic/chemistry , Administration, Intranasal , Animals , Mice , Mice, Inbred BALB C , Saponins/chemistry , Spectrum Analysis/methods
11.
J Infect Dis ; 214(10): 1588-1596, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27112503

ABSTRACT

For many bacterial respiratory infections, development of (severe) disease is preceded by asymptomatic colonization of the upper airways. For Streptococcus pneumoniae, the transition to severe lower respiratory tract infection is associated with an increase in nasopharyngeal colonization density. Insight into how the mucosal immune system restricts colonization may provide new strategies to prevent clinical symptoms. Several studies have provided indirect evidence that the mucosal adjuvant cholera toxin subunit B (CTB) may confer nonspecific protection against respiratory infections. Here, we show that CTB reduces the pneumococcal load in the nasopharynx, which required activation of the caspase-1/11 inflammasome, mucosal T cells, and macrophages. Our findings suggest that CTB-dependent activation of the local innate response synergizes with noncognate T cells to restrict bacterial load. Our study not only provides insight into the immunological components required for containment and clearance of pneumococcal carriage, but also highlights an important yet often understudied aspect of adjuvants.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antigens, Bacterial/analysis , Bacterial Load , Carrier State/immunology , Cholera Toxin/pharmacology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/isolation & purification , Adjuvants, Immunologic/administration & dosage , Administration, Mucosal , Animals , Antigens , Cholera Toxin/administration & dosage , Inflammasomes/metabolism , Macrophages/immunology , Mice, Inbred C57BL , Nasopharynx/microbiology , Streptococcus pneumoniae/immunology , T-Lymphocytes/immunology
12.
Microbiol Immunol ; 60(7): 497-500, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27301339

ABSTRACT

In this study, the mucosal adjuvant activity of Salmonella flagellin as a carrier in a conjugate of EXP153-rFliC was investigated. EXP153-rFliC was made by conjugation of a synthetic B-cell epitope peptide derived from Plasmodium falciparum exported protein-1(EXP153) to recombinant phase 1 flagellin of Salmonella enterica serovar Typhimurium expressed in Escherichia coli (rFliC), and used to immunize BALB/c mice via intranasal instillation. It was found that robust EXP153-specific serum IgG antibodies were induced without additional adjuvant. EXP153-specific sIgA antibodies were also induced, these being detected in bronchoalveolar, nasal, vaginal and intestinal washes. These observations demonstrate that Salmonella flagellin as a carrier is an effective mucosal adjuvant in that its conjugated peptide induces antibody responses.


Subject(s)
Antibodies, Bacterial/immunology , Antibody Formation/immunology , Flagellin/immunology , Immunity, Mucosal , Nasal Mucosa/immunology , Peptide Fragments/immunology , Salmonella typhimurium/immunology , Administration, Intranasal , Animals , Antibody Specificity/immunology , Female , Flagellin/chemistry , Immunoglobulin A, Secretory/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Mice , Peptide Fragments/administration & dosage
13.
Int J Mol Sci ; 17(9)2016 Aug 27.
Article in English | MEDLINE | ID: mdl-27618897

ABSTRACT

The nontoxic heat-labile toxin (LT) B subunit (LTB) was used as mucosal adjuvant experimentally. However, the mechanism of LTB adjuvant was still unclear. The LTB and enterovirus 71 (EV71) VP1 subunit (EVP1) were constructed in pET32 and expressed in E. coli BL21, respectively. The immunogenicity of purified EVP1 and the adjuvanticity of LTB were evaluated via intranasal immunization EVP1 plus LTB in Balb/c mice. In order to elucidate the proteome change triggered by the adjuvant of LTB, the proteomic profiles of LTB, EVP1, and LTB plus EVP1 were quantitatively analyzed by iTRAQ-LC-MS/MS (isobaric tags for relative and absolute quantitation; liquid chromatography-tandem mass spectrometry) in murine macrophage RAW264.7. The proteomic data were analyzed by bioinformatics and validated by western blot analysis. The predicted protein interactions were confirmed using LTB pull-down and the LTB processing pathway was validated by confocal microscopy. The results showed that LTB significantly boosted EVP1 specific systematic and mucosal antibodies. A total of 3666 differential proteins were identified in the three groups. Pathway enrichment of proteomic data predicted that LTB upregulated the specific and dominant MAPK (mitogen-activated protein kinase) signaling pathway and the protein processing in endoplasmic reticulum (PPER) pathway, whereas LTB or EVP1 did not significantly upregulate these two signaling pathways. Confocal microscopy and LTB pull-down assays confirmed that the LTB adjuvant was endocytosed and processed through endocytosis (ENS)-lysosomal-endoplasmic reticulum (ER) system.


Subject(s)
Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Enterotoxins/immunology , Enterotoxins/metabolism , Enterovirus A, Human/metabolism , Escherichia coli Proteins/immunology , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Proteomics/methods , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/metabolism , Animals , Antibodies, Viral/immunology , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Enterotoxins/chemistry , Enterotoxins/genetics , Enterovirus A, Human/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Mice , Microscopy, Confocal
14.
Clin Immunol ; 161(2): 251-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26111481

ABSTRACT

EP67 is a complement component 5a (C5a)-derived peptide agonist of the C5a receptor (CD88) that selectively activates DCs over neutrophils. Systemic administration of EP67 covalently attached to peptides, proteins, or attenuated pathogens generates TH1-biased immunogen-specific humoral and cellular immune responses with little inflammation. Furthermore, intranasal administration of EP67 alone increases the proportion of activated APCs in the airways. As such, we hypothesized that EP67 can act as a mucosal adjuvant. Intranasal immunization with an EP67-conjugated CTL peptide vaccine against protective MCMV epitopes M84 and pp89 increased protection of naïve female BALB/c mice against primary respiratory infection with salivary gland-derived MCMV and generated higher proportions of epitope responsive and long-lived memory precursor effector cells (MPEC) in the lungs and spleen compared to an inactive, scrambled EP67-conjugated CTL peptide vaccine and vehicle alone. Thus, EP67 may be an effective adjuvant for mucosal vaccines and warrants further study.


Subject(s)
Herpesviridae Infections/immunology , Immunity, Mucosal/immunology , Muromegalovirus/immunology , Oligopeptides/immunology , Vaccines, Subunit/immunology , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Amino Acid Sequence , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Epitopes/immunology , Female , Flow Cytometry , Herpesviridae Infections/prevention & control , Herpesviridae Infections/virology , Host-Pathogen Interactions/immunology , Immunity, Mucosal/drug effects , Lung/immunology , Lung/virology , Mice , Mice, Inbred BALB C , Muromegalovirus/physiology , NIH 3T3 Cells , Oligopeptides/administration & dosage , Salivary Glands/immunology , Salivary Glands/virology , Spleen/immunology , Spleen/virology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , Vaccination/methods , Vaccines, Subunit/administration & dosage
15.
Int J Biol Macromol ; 281(Pt 1): 136208, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39362439

ABSTRACT

OBJECTIVES: Recently, more and more evidences suggest that ß-glucans can induce trained immunity and non-specific protections against pathogens. However, most of the reports evaluated the immunological activities of ß-glucans through injection route but no nasal inhalation. In this study, the effects of curdlan sulfate-based nanoparticles, CS/O-HTCC on trained immunity through intranasal administration were evaluated. METHODS: Macrophages were treated with CS/O-HTCC and the metabolisms of the macrophages were detected. Mice were intranasal administered with CS/O-HTCC for 3 times with a 14 days interval, then the antitumor or infection prevention effects were assessed. RESULTS: In vitro, CS/O-HTCC enhanced the macrophage metabolism significantly through upregulating glycolysis (26.1 ± 4.3 mpH/min) and oxidative phosphorylation (36.0 ± 9.0 pmol/min) compared with that of negative group (7.5 ± 2.3 mpH/min and 19.5 ± 4.9 pmol/min). In vivo, CS/O-HTCC inhibited lung metastasis of B16F10 tumor cells and improved the survival time (26.5 days) of the nmice compared with negative group (19.5 days). Moreover, CS/O-HTCC prevented the lung infections by Escherichia coli or Streptococcus pneumoniae (less bacterial residual) and reduced lung damages. CONCLUSIONS: CS/O-HTCC can induce trained immunity through enhancing the metabolism of macrophages and enhance the non-specific protection against pathogens through intranasal immunization.

16.
Vaccines (Basel) ; 12(5)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38793700

ABSTRACT

The development of mucosal vaccines, which can generate antigen-specific immune responses in both the systemic and mucosal compartments, has been recognized as an effective strategy for combating infectious diseases caused by pathogenic microbes. Our recent research has focused on creating a nasal vaccine system in mice using enzymatically polymerized caffeic acid (pCA). However, we do not yet understand the molecular mechanisms by which pCA stimulates antigen-specific mucosal immune responses. In this study, we hypothesized that pCA might activate mucosal immunity at the site of administration based on our previous findings that pCA possesses immune-activating properties. However, contrary to our initial hypothesis, the intranasal administration of pCA did not enhance the expression of various genes involved in mucosal immune responses, including the enhancement of IgA responses. Therefore, we investigated whether pCA forms a complex with antigenic proteins and enhances antigen delivery to mucosal dendritic cells located in the lamina propria beneath the mucosal epithelial layer. Data from gel filtration chromatography indicated that pCA forms a complex with the antigenic protein ovalbumin (OVA). Furthermore, we examined the promotion of OVA delivery to nasal mucosal dendritic cells (mDCs) after the intranasal administration of pCA in combination with OVA and found that OVA uptake by mDCs was increased. Therefore, the data from gel filtration chromatography and flow cytometry imply that pCA enhances antigen-specific antibody production in both mucosal and systemic compartments by serving as an antigen-delivery vehicle.

17.
Int J Biol Macromol ; 266(Pt 2): 131289, 2024 May.
Article in English | MEDLINE | ID: mdl-38570002

ABSTRACT

Intranasal vaccination offers crucial protection against influenza virus pandemics. However, antigens, especially subunit antigens, often fail to induce effective immune responses without the help of immune adjuvants. Our research has demonstrated that a polyelectrolyte complex, composed of curdlan sulfate/O-(2-hydroxyl) propyl-3-trimethyl ammonium chitosan chloride (CS/O-HTCC), effectively triggers both mucosal and systemic immune responses when administrated intranasal. In this study, stable nanoparticles formed by curdlan-O-HTCC conjugate (CO NP) were prepared and characterized. Furthermore, the efficacy of CO NP was evaluated as a mucosal adjuvant in an intranasal influenza H1N1 subunit vaccine. The results revealed that CO NP exhibits uniform and spherical morphology, with a size of 190.53 ± 4.22 nm, and notably, it remains stable in PBS at 4 °C for up to 6 weeks. Biological evaluation demonstrated that CO NP stimulates the activation of antigen-presenting cells (APCs), including macrophages and dendritic cells (DCs), both in vitro and in vivo. Furthermore, intranasal administration of CO NP effectively elicits cellular and humoral immune responses, notably enhancing mucosal immunity. Thus, CO NP emerges as a promising mucosal adjuvant for influenza subunit vaccines.


Subject(s)
Adjuvants, Immunologic , Administration, Intranasal , Chitosan , Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Nanoparticles , Vaccines, Subunit , beta-Glucans , Influenza A Virus, H1N1 Subtype/immunology , Chitosan/chemistry , Nanoparticles/chemistry , Influenza Vaccines/immunology , Influenza Vaccines/chemistry , Influenza Vaccines/administration & dosage , beta-Glucans/chemistry , beta-Glucans/pharmacology , beta-Glucans/administration & dosage , Animals , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/administration & dosage , Mice , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Immunity, Mucosal/drug effects , Mice, Inbred BALB C , Female , Dendritic Cells/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology
18.
Hum Vaccin Immunother ; 20(1): 2374147, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39090779

ABSTRACT

Entamoeba histolytica, the causative agent of amebiasis, is one of the top three parasitic causes of mortality worldwide. However, no vaccine exists against amebiasis. Using a lead candidate vaccine containing the LecA fragment of Gal-lectin and GLA-3M-052 liposome adjuvant, we immunized rhesus macaques via intranasal or intramuscular routes. The vaccine elicited high-avidity functional humoral responses as seen by the inhibition of amebic attachment to mammalian target cells by plasma and stool antibodies. Importantly, antigen-specific IFN-γ-secreting peripheral blood mononuclear cells (PBMCs) and IgG/IgA memory B cells (BMEM) were detected in immunized animals. Furthermore, antigen-specific antibody and cellular responses were maintained for at least 8 months after the final immunization as observed by robust LecA-specific BMEM as well as IFN-γ+ PBMC responses. Overall, both intranasal and intramuscular immunizations elicited a durable and functional response in systemic and mucosal compartments, which supports advancing the LecA+GLA-3M-052 liposome vaccine candidate to clinical testing.


Subject(s)
Administration, Intranasal , Antibodies, Protozoan , Entamoeba histolytica , Entamoebiasis , Interferon-gamma , Leukocytes, Mononuclear , Liposomes , Macaca mulatta , Protozoan Vaccines , Animals , Entamoeba histolytica/immunology , Liposomes/immunology , Liposomes/administration & dosage , Protozoan Vaccines/immunology , Protozoan Vaccines/administration & dosage , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Leukocytes, Mononuclear/immunology , Entamoebiasis/prevention & control , Entamoebiasis/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Injections, Intramuscular , Immunogenicity, Vaccine , Adjuvants, Vaccine/administration & dosage , Adjuvants, Immunologic/administration & dosage , B-Lymphocytes/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin A/immunology , Immunoglobulin A/blood , Antigens, Protozoan/immunology , Immunity, Humoral , Immunologic Memory , Protozoan Proteins/immunology
19.
Life Sci ; 355: 122986, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39151885

ABSTRACT

Brucellosis is a chronic infectious disease that is zoonotic in nature. Brucella can infect humans through interactions with livestock, primarily via the digestive tract, respiratory tract, and oral cavity. This bacterium has the potential to be utilized as a biological weapon and is classified as a Category B pathogen by the Centers for Disease Control and Prevention. Currently, there is no approved vaccine for humans against Brucella, highlighting an urgent need for the development of a vaccine to mitigate the risks posed by this pathogen. Brucella primarily infects its host by adhering to and penetrating mucosal surfaces. Mucosal immunity plays a vital role in preventing local infections, clearing microorganisms from mucosal surfaces, and inhibiting the spread of pathogens. As mucosal vaccine strategies continue to evolve, the development of a safe and effective mucosal vaccine against Brucella appears promising.This paper reviews the immune mechanism of mucosal vaccines, the infection mechanism of Brucella, successful Brucella mucosal vaccines in animals, and mucosal adjuvants. Additionally, it elucidates targeting and optimization strategies for mucosal vaccines to facilitate the development of human vaccines against Brucella.


Subject(s)
Brucella Vaccine , Brucella , Brucellosis , Immunity, Mucosal , Humans , Animals , Brucella/immunology , Immunity, Mucosal/immunology , Brucellosis/prevention & control , Brucellosis/immunology , Brucellosis/microbiology , Brucella Vaccine/immunology , Adjuvants, Immunologic , Vaccine Development
20.
Vaccines (Basel) ; 12(4)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38675794

ABSTRACT

Previously, it was shown that intranasally (i.n.) administered Corynebacterium pseudodiphtheriticum 090104 (Cp) or CP-derived bacterium-like particles (BLPs) improve the immunogenicity of the pneumococcal conjugate vaccine (PCV). This work aimed to deepen the characterization of the adjuvant properties of Cp and CP-derived BLPs for their use in the development of pneumococcal vaccines. The ability of Cp and CP-derived BLPs to improve both the humoral and cellular specific immune responses induced by i.n. administered polysaccharide-based commercial pneumococcal vaccine (Pneumovax 23®) and the chimeric recombinant PSPF (PsaA-Spr1875-PspA-FliC) protein was evaluated, as well as the protection against Streptococcus pneumoniae infection in infant mice. Additionally, whether the immunization protocols, including Cp and CP-derived BLPs, together with the pneumococcal vaccines can enhance the resistance to secondary pneumococcal pneumonia induced after inflammatory lung damage mediated by the activation of Toll-like receptor 3 (TLR3) was assessed. The results showed that both Cp and CP-derived BLPs increased the immunogenicity and protection induced by two pneumococcal vaccines administered through the nasal route. Of note, the nasal priming with the PSPF T-dependent antigen co-administered with Cp or CP-derived BLPs efficiently stimulated humoral and cellular immunity and increased the resistance to primary and secondary pneumococcal infections. The CP-derived BLPs presented a stronger effect than live bacteria. Given safety concerns associated with live bacterium administration, especially in high-risk populations, such as infants, the elderly, and immunocompromised patients, BLPs emerge as an attractive mucosal adjuvant to improve the host response to pneumococcal infections and to enhance the vaccines already in the market or in development.

SELECTION OF CITATIONS
SEARCH DETAIL