Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
NPJ Vaccines ; 8(1): 120, 2023 Aug 14.
Article in English | MEDLINE | ID: mdl-37580335

ABSTRACT

The mucosal delivery route is considered ideal for immunization. However, induction of antigen-specific mucosal immunity is difficult due to the tolerogenic environment. Therefore, developing an immunogenic mucosal dendritic cell (DC)-targeting strategy is required. Herein, we investigated the characteristics and immunogenic potential of Peyer's patch (PP) DCs as an oral vaccination-targeting strategy. Single-cell RNA sequencing analysis of the PP DCs showed that complement C5a receptor- and lysozyme-expressing DCs exhibit increased expression of genes related to chemotaxis. Administration of the Co1 peptide, a C5aR ligand, increased CD8+ T cell infiltration and response to the co-delivered model antigen in mice. Furthermore, in the SARS-CoV-2 vaccine model, vaccination with Co1 elicited both systemic and mucosal immunity. Collectively, these findings demonstrate that C5aR signaling in mucosal DCs plays a role in regulating adjuvant activity by modulating the tissue microenvironment.

2.
Cell Rep ; 35(2): 108995, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33852847

ABSTRACT

The complement fragment C5a is closely associated with adaptive immune induction in the mucosa. However, the mechanisms that control CD8+ T cell responses by C5a have not been extensively explored. This study reveals that C5/C5a in the Peyer's patch (PP) subepithelial dome increases upon oral Listeria infection. We hypothesize that C5aR+ PP cells play an important role in the induction of antigen-specific T cell immunity. Using single-cell RNA sequencing, we identify C5aR- and lysozyme-expressing dendritic cells (C5aR+ LysoDCs) in PP and examine their role in CD8+ T cell immune induction. Stimulation of C5aR+ LysoDCs by C5a increases reactive oxygen species levels, leading to efficient antigen cross-presentation, which elicits an antigen-specific CD8+ T cell response. In C5-deficient mice, oral co-administration of C5a and Listeria enhances Listeria-specific cytotoxic T cell levels. Collectively, these findings suggest a role of the complement system in intestinal T cell immunity.


Subject(s)
Complement C5a/immunology , Cross-Priming , Intestinal Mucosa/immunology , Listeria monocytogenes/immunology , Peyer's Patches/immunology , Receptor, Anaphylatoxin C5a/genetics , T-Lymphocytes, Cytotoxic/immunology , Adaptive Immunity , Animals , Antigen Presentation , Complement C5a/genetics , Complement C5a/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/microbiology , Female , Gene Expression Profiling , Gene Expression Regulation , Immunity, Mucosal , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Listeria monocytogenes/pathogenicity , Macrophages/drug effects , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/immunology , Monocytes/microbiology , Muramidase/genetics , Muramidase/immunology , Peyer's Patches/drug effects , Peyer's Patches/microbiology , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Receptor, Anaphylatoxin C5a/immunology , Single-Cell Analysis , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/microbiology
3.
Immune Netw ; 20(6): e44, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33425429

ABSTRACT

The human body is continuously threatened by pathogens, and the immune system must maintain a balance between fighting infection and becoming over-activated. Mucosal surfaces cover several anatomically diverse organs throughout the body, such as the respiratory and gastrointestinal tracts, and are directly exposed to the external environment. Various pathogens invade the body through mucosal surfaces, making the mucosa the frontline of immune defense. The immune systems of various mucosal tissues display distinctive features that reflect the tissues' anatomical and functional characteristics. This review discusses the cellular components that constitute the respiratory and gastrointestinal tracts; in particular, it highlights the complex interactions between epithelial and immune cells to induce Ag-specific immune responses in the lung and gut. This information on mucosal immunity may facilitate understanding of the defense mechanisms against infectious agents that invade mucosal surfaces, such as severe acute respiratory syndrome coronavirus 2, and provide insight into effective vaccine development.

4.
Immune Netw ; 18(3): e21, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29984039

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) is a contagious coronavirus infecting pigs that leads to significant economic losses in the swine industry. Given that PEDV infection occurs in gut epithelial cells mainly via the fecal-oral route, induction of PEDV-specific immune responses in the mucosal compartment is required for protective immunity against viral infection. However, an effective mucosal vaccine against the currently prevalent PEDV strain is not available. In this study, we demonstrated that the N-terminal domain (NTD) of the spike (S) protein of PEDV represents a new vaccine candidate molecule to be applied via the mucosal route. We first established an Escherichia coli expression system producing the partial NTD (NTD231-501) of the PEDV S protein. Orally administered NTD231-501 protein specifically interacted with the apical area of M cells in the follicle-associated epithelium of Peyer's patch. Additionally, the NTD protein induced antigen-specific immune responses in both the systemic and mucosal immune compartments when administered orally. Collectively, we propose the NTD of the PEDV S protein to be a candidate mucosal vaccine molecule.

5.
Cell Immunol ; 325: 41-47, 2018 03.
Article in English | MEDLINE | ID: mdl-29397905

ABSTRACT

Mucosal vaccination is an ideal strategy to induce protective immunity in both mucosal and parenteral areas. Successful induction of an antigen-specific immune response via mucosal administration essentially requires the effective delivery of antigen into a mucosal immune inductive site, which depends on antigen delivery into M cells. We previously reported that M cells specifically express C5aR, and antigen targeting to C5aR by using specific ligands, including Co1 peptide, promotes the antigen-specific immune response in both mucosal and systemic immune compartments. In this study, we found that application of the Co1 peptide to dengue virus antigen containing CD8 T cell epitopes effectively induced an antigen-specific IFN-γ-producing CD8+ T cell response after oral mucosal administration of antigen. Consequently, we suggest that Co1 peptide-mediated C5aR targeting of antigen into M cells can be used for the induction of an effective antigen-specific CD8+ T cell immune response in oral mucosal vaccine development.


Subject(s)
Dengue Vaccines/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Viral Nonstructural Proteins/immunology , Animals , Antigens , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Dengue Virus/metabolism , Disease Models, Animal , Immunity, Mucosal/immunology , Mice , Mice, Inbred BALB C , Mucous Membrane/immunology , Vaccination , Viral Nonstructural Proteins/metabolism
6.
Immune Netw ; 17(4): 228-236, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28860952

ABSTRACT

In the intestinal mucosal surface, microfold cells (M cells) are the representative gateway for the uptake of luminal antigens. At the same time, M cells are the primary infection site for pathogens invading mucosal surface for their infection. Although it is well recognized that many mucosal pathogens exploit the M cells for their infection, the mechanism to infect M cells utilized by pathogens is not clearly understood yet. In this study, we found that M cells expressing complement 5a (C5a) receptor (C5aR) also express Toll-like receptor (TLR) 1/2 and TLR4. Infection of Yersinia enterocolitica, an M cell-invading pathogen, synergistically regulated cyclic adenosine monophosphate-dependent protein kinase A (cAMP-PKA) signaling which are involved in signal crosstalk between C5aR and TLRs. In addition, Y. enterocolitica infection into M cells was enhanced by C5a treatment and this enhancement was abrogated by C5a antagonist treatment. Finally, Y. enterocolitica infection into M cells was unsuccessful in C5aR knock-out mice. Collectively, we suggest that exploit the crosstalk between C5aR and TLR signaling is one of infection mechanisms utilized by mucosal pathogens to infect M cells.

7.
Sci Rep ; 7(1): 3980, 2017 06 21.
Article in English | MEDLINE | ID: mdl-28638068

ABSTRACT

The regional specialization of intestinal immune cells is affected by the longitudinal heterogeneity of environmental factors. Although the distribution of group 3 innate lymphoid cells (ILC3s) is well characterized in the lamina propria, it is poorly defined in Peyer's patches (PPs) along the intestine. Given that PP ILC3s are closely associated with mucosal immune regulation, it is important to characterize the regulatory mechanism of ILC3s. Here, we found that terminal ileal PPs of specific pathogen-free (SPF) mice have fewer NKp46+ ILC3s than jejunal PPs, while there was no difference in NKp46+ ILC3 numbers between terminal ileal and jejunal PPs in antibiotics (ABX)-treated mice. We also found that butyrate levels in the terminal ileal PPs of SPF mice were higher than those in the jejunal PPs of SPF mice and terminal ileal PPs of ABX-treated mice. The reduced number of NKp46+ ILC3s in terminal ileal PPs resulted in a decrease in Csf2 expression and, in turn, resulted in reduced regulatory T cells and enhanced antigen-specific T-cell proliferation. Thus, we suggest that NKp46+ ILC3s are negatively regulated by microbiota-derived butyrate in terminal ileal PPs and the reduced ILC3 frequency is closely associated with antigen-specific immune induction in terminal ileal PPs.


Subject(s)
Butyrates/administration & dosage , Gastrointestinal Microbiome , Ileum/immunology , Jejunum/immunology , Lymphocytes/microbiology , Peyer's Patches/immunology , Animals , Antigens, Ly/metabolism , Butyrates/analysis , Ileum/microbiology , Jejunum/microbiology , Lymphocytes/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Natural Cytotoxicity Triggering Receptor 1/metabolism , Peyer's Patches/microbiology
8.
Chin J Nat Med ; 15(3): 192-201, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28411687

ABSTRACT

Red ginseng is a well-known alternative medicine with anti-inflammatory activity. It exerts pharmacological effects through the transformation of saponin into metabolites by intestinal microbiota. Given that intestinal microflora vary among individuals, the pharmacological effects of red ginseng likely vary among individuals. In order to produce homogeneously effective red ginseng, we prepared probiotic-fermented red ginseng and evaluated its activity using a dextran sulfate sodium (DSS)-induced colitis model in mice. Initial analysis of intestinal damage indicated that the administration of probiotic-fermented red ginseng significantly decreased the severity of colitis, compared with the control and the activity was higher than that induced by oral administration of ginseng powder or probiotics only. Subsequent analysis of the levels of serum IL-6 and TNF-α, inflammatory biomarkers that are increased at the initiation stage of colitis, were significantly decreased in probiotic-fermented red ginseng-treated groups in comparison to the control group. The levels of inflammatory cytokines and mRNAs for inflammatory factors in colorectal tissues were also significantly decreased in probiotic-fermented red ginseng-treated groups. Collectively, oral administration of probiotic-fermented red ginseng reduced the severity of colitis in a mouse model, suggesting that it can be used as a uniformly effective red ginseng product.


Subject(s)
Colitis/drug therapy , Lactobacillus plantarum/metabolism , Panax/microbiology , Plant Extracts/administration & dosage , Probiotics/metabolism , Administration, Oral , Animals , Colitis/chemically induced , Colitis/immunology , Colon/drug effects , Colon/immunology , Dextran Sulfate/adverse effects , Disease Models, Animal , Female , Fermentation , Humans , Interleukin-6/immunology , Mice , Mice, Inbred BALB C , Panax/chemistry , Panax/metabolism , Plant Extracts/chemistry , Plant Extracts/metabolism , Powders/administration & dosage , Powders/metabolism , Tumor Necrosis Factor-alpha/immunology
9.
Clin Exp Vaccine Res ; 6(1): 15-21, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28168169

ABSTRACT

Vaccination is the most successful immunological practice that improves the quality of human life and health. Vaccine materials include antigens of pathogens and adjuvants potentiating the effectiveness of vaccination. Vaccines are categorized using various criteria, including the vaccination material used and the method of administration. Traditionally, vaccines have been injected via needles. However, given that most pathogens first infect mucosal surfaces, there is increasing interest in the establishment of protective mucosal immunity, achieved by vaccination via mucosal routes. This review summarizes recent developments in mucosal vaccines and their associated adjuvants.

10.
J Immunol ; 198(2): 629-633, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27974458

ABSTRACT

Peyer's patches (PPs) are the major mucosal immune-inductive site, and germinal centers (GCs) in PPs determine the quality of the Abs produced. PP GCs are continuously induced by the gut microbiota, and their maintenance contributes to the induction of strong IgA responses to Ags. In this study, we investigated the role of formyl peptide receptor (FPR)-mediated signaling in the maintenance of PP GCs, because FPRs recognize the microbiota and initiate an innate immune response by chemotaxis. We found that follicular dendritic cells (FDCs), a key organizer of B cell follicles and GCs in mucosal immunity, express Fpr2. Additionally, Fpr2-mediated signaling in PP FDCs promoted Cxcl13 and B cell activating factor expression, as well as B cell proliferation and activation. Therefore, we suggest that Fpr2-mediated signaling in FDCs plays a key role in GC maintenance in PPs and results in an Ag-specific IgA response in the gut mucosal immune compartment.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells, Follicular/immunology , Lymphocyte Activation/immunology , Peyer's Patches/immunology , Receptors, Formyl Peptide/immunology , Animals , Antimicrobial Cationic Peptides/immunology , Flow Cytometry , Fluorescent Antibody Technique , Germinal Center/immunology , Immunity, Mucosal/immunology , Mice , Mice, Inbred BALB C , Real-Time Polymerase Chain Reaction , Signal Transduction/immunology , Cathelicidins
11.
Pharm Biol ; 55(1): 317-323, 2017 Dec.
Article in English | MEDLINE | ID: mdl-27927080

ABSTRACT

CONTEXT: Red ginseng (heat-processed Panax ginseng) is a well-known alternative medicine with pharmacological antidiabetic activity. It exerts pharmacological effects through the transformation of saponin into metabolites by the intestinal microbiota. Given that intestinal conditions and intestinal microflora vary among individuals, the pharmacological effects of orally administered red ginseng likely may vary among individuals. OBJECTIVE: To overcome this variation and produce homogeneously effective red ginseng, we evaluated the antidiabetic effects of probiotic-fermented red ginseng in a mouse model. MATERIALS AND METHODS: The antidiabetic efficacy of orally administered probiotic-fermented red ginseng was assessed in ICR mice after induction of diabetes using streptozotocin (170 mg/kg body weight). Samples were given orally for 8 weeks, and indicators involved in diabetic disorders such as body weight change, water intake, blood glucose, glucose tolerance and various biochemical parameters were determined. RESULTS: Oral administration of probiotic-fermented red ginseng significantly decreased the level of blood glucose of about 62.5% in the fasting state and induced a significant increase in glucose tolerance of about 10.2% compared to the control diabetic mice. Additionally, various indicators of diabetes and biochemical data (e.g., blood glycosylated haemoglobin level, serum concentrations of insulin, and α-amylase activity) showed a significant improvement in the diabetic conditions of the mice treated with probiotic-fermented red ginseng in comparison with those of control diabetic mice. DISCUSSION AND CONCLUSION: Our results demonstrate the antidiabetic effects of probiotic-fermented red ginseng in the streptozotocin-induced mouse diabetes model and suggest that probiotic-fermented red ginseng may be a uniformly effective red ginseng product.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Experimental/drug therapy , Fermentation , Hypoglycemic Agents/pharmacology , Panax/metabolism , Plant Extracts/metabolism , Plant Extracts/pharmacology , Probiotics , Streptozocin , Administration, Oral , Animals , Biomarkers/blood , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/chemically induced , Glycated Hemoglobin/metabolism , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/metabolism , Insulin/blood , Male , Mice, Inbred ICR , Phytotherapy , Plant Extracts/administration & dosage , Plants, Medicinal , Powders , Time Factors , alpha-Amylases/metabolism
12.
Biochem Biophys Res Commun ; 473(4): 894-898, 2016 05 13.
Article in English | MEDLINE | ID: mdl-27059141

ABSTRACT

Dengue virus (DENV) is a mosquito-borne pathogen that annually infects more than 390 million people in 100 different countries. Symptoms of the viral infection include a relatively weak dengue fever to severe dengue hemorrhagic fever/dengue shock syndrome, which are mortal infectious diseases. As of yet, there is no commercially available vaccine or therapeutic for DENV. Currently, passive immunotherapy using DENV-specific antibody (Ab) is a considered strategy to treat DENV infection. Here, we developed a monoclonal Ab (mAb), EDIIImAb-61, specific to the DENV domain III of the envelope glycoprotein (EDIII) with broad-spectrum detection ability to all four DENV serotypes (DENV-1∼4) to use as a therapeutic Ab. Although EDIII contains non-immunodominant epitopes compared to domains I and II, domain III plays a critical role in host receptor binding. EDIIImAb-61 exhibited cross-reactive binding affinity to all four DENV serotypes that had been isolated from infected humans. To further characterize EDIIImAb-61 and prepare genes for large-scale production using a heterologous expression system, the sequence of the complementarity determining regions was analyzed after cloning the full-length cDNA genes encoding the heavy and light chain of the mAb. Finally, we produced Ab from CHO-K1 cells transfected with the cloned EDIIImAb-61 heavy and light chain genes and confirmed the binding ability of the Ab. Collectively, we conclude that EDIIImAb-61 itself and the recombinant Ab produced using the cloned heavy and light chain gene of EDIIImAb-61 is a candidate for passive immunotherapy against DENV infection.


Subject(s)
Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Dengue Virus/immunology , Viral Envelope Proteins/metabolism , Animals , Binding Sites , CHO Cells , Cricetulus , Dengue Virus/genetics , Dengue Virus/isolation & purification , Drug Design , Protein Binding , Protein Engineering , Serogroup , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
13.
Vet Microbiol ; 177(1-2): 142-53, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25818577

ABSTRACT

Actinobacillus pleuropneumoniae is the causative agent of porcine pleuropneumonia and severe economic loss in the swine industry has been caused by the infection. Therefore, the development of an effective vaccine against the bacteria is necessary. ApxII toxin, among several virulence factors expressed by the bacteria, is considered to be a promising vaccine candidate because ApxII toxin not only accompanies cytotoxic and hemolytic activities, but is also expressed in all 15 serotypes of bacteria except serotypes 10 and 14. In this study, we identified the peptide ligand capable of targeting the ligand-conjugated ApxIIA #5 fragment antigen to nasopharynx-associated lymphoid tissue. It was found that nasal immunization with ligand-conjugated ApxIIA #5 induced efficient mucosal and systemic immune responses measured at the levels of antigen-specific antibodies, cytokine-secreting cells after antigen exposure, and antigen-specific lymphocyte proliferation. More importantly, the nasal immunization induced protective immunity against nasal challenge infection of the bacteria, which was confirmed by histopathological studies and bacterial clearance after challenge infection. Collectively, we confirmed that the ligand capable of targeting the ligand-conjugated antigen to nasopharynx-associated lymphoid tissue can be used as an effective nasal vaccine adjuvant to induce protective immunity against A. pleuropneumoniae infection.


Subject(s)
Actinobacillus Infections/veterinary , Actinobacillus pleuropneumoniae/immunology , Bacterial Vaccines/immunology , Immunization/veterinary , Pleuropneumonia/veterinary , Swine Diseases/microbiology , Swine Diseases/prevention & control , Actinobacillus Infections/microbiology , Actinobacillus Infections/prevention & control , Actinobacillus pleuropneumoniae/pathogenicity , Administration, Intranasal , Animals , Bacterial Vaccines/administration & dosage , Mice , Pleuropneumonia/immunology , Pleuropneumonia/prevention & control , Swine
14.
Eur J Immunol ; 45(5): 1402-13, 2015 May.
Article in English | MEDLINE | ID: mdl-25655317

ABSTRACT

The human antimicrobial peptide LL-37 is known to have chemotactic and modulatory activities on various cells including monocytes, T cells, and epithelial cells. Given that LL-37 enhances chemotactic attraction and modulates the activity of DCs, it is conceivable that it might play a role as an immune adjuvant by skewing the immune environment toward immunostimulatory conditions. In this study, we characterized the mucosal adjuvant activity of LL-37 using model and pathogenic Ags. When LL-37-conjugated Ag was administered orally to mice, a tolerogenic Peyer's patch environment was altered to cell populations containing IL-6-secreting CD11c(+), CD11c(+) CD70(+), and Th17 cells capable of evoking a subsequent LL-37-conjugated Ag-specific immune response in both systemic and mucosal immune compartments. In addition, we showed presentation of formyl peptide receptor, an LL-37 receptor, on M cells, which may aid the initiation of an LL-37-mediated enhanced immune response through targeting and transcytosis of the conjugated Ag. Based on our findings, we conclude that LL-37 has potential as an oral mucosal adjuvant, not only by enhancing the delivery of LL-37-conjugated Ag to M cells, but also by triggering T-cell-mediated Ag-specific immune responses through modulation of the mucosal immune environment.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cathelicidins/administration & dosage , Cathelicidins/immunology , Immunity, Mucosal , Th17 Cells/immunology , Administration, Oral , Animals , Antibodies, Neutralizing/biosynthesis , Antimicrobial Cationic Peptides , Cytokines/biosynthesis , Dengue Vaccines/immunology , Female , Green Fluorescent Proteins/administration & dosage , Green Fluorescent Proteins/immunology , Humans , Immunoglobulin A, Secretory/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Peyer's Patches/cytology , Peyer's Patches/immunology
15.
Immune Netw ; 15(1): 44-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25713508

ABSTRACT

Interactions between microbes and epithelial cells in the gastrointestinal tract are closely associated with regulation of intestinal mucosal immune responses. Recent studies have highlighted the modulation of mucosal immunity by microbe-derived molecules such as ATP and short-chain fatty acids. In this study, we undertook to characterize the expression of the ATP-gated P2X7 receptor (P2X7R) on M cells and its role in gastrointestinal mucosal immune regulation because it was poorly characterized in Peyer's patches, although purinergic signaling via P2X7R and luminal ATP have been considered to play an important role in the gastrointestinal tract. Here, we present the first report on the expression of P2X7R on M cells and characterize the role of P2X7R in immune enhancement by ATP or LL-37.

16.
Int J Med Mushrooms ; 17(12): 1189-99, 2015.
Article in English | MEDLINE | ID: mdl-26854106

ABSTRACT

Ophiocordyceps sinensis is a natural fungus that has been valued as a health food and traditional Chinese medicine for centuries. The fungus is parasitic and colonizes insect larva. Naturally occurring O. sinensis thrives at high altitude in cold and grassy alpine meadows on the Himalayan mountain ranges. Wild O. sinensis is becoming increasingly rare in its natural habitats, and its price is out of reach for clinical practice. For these reasons, development of a standardized alternative is a great focus of research to allow the use of O. sinensis as a medicine. To develop an alternative for wild O. sinensis, a refined standardized extract, CBG-CS-2, was produced by artificial fermentation and extraction of the mycelial strain Paecilomyces hepiali CBG-CS-1, which originated from wild O. sinensis. In this study, we analyzed the in vivo immune-modulating effect of CBG-CS-2 in mice. Oral administration of CBG-CS-2 supported splenocyte stimulation and enhanced Th1-type cytokine expression from the splenocytes. Importantly, the same treatment significantly enhanced the natural killer cell activity of the splenocytes. Finally, oral administration of CBG-CS-2 enhanced the potential for inflammatory responses. Together, these findings indicate that the mycelial culture extract prepared from O. sinensis exhibited immune-modulating activity and suggest its possible use in the treatment of diseases caused by abnormal immune function.


Subject(s)
Ascomycota/chemistry , Biological Products/isolation & purification , Immunologic Factors/pharmacology , Pharmaceutical Preparations/isolation & purification , Animals , Biological Products/pharmacology , Cell Proliferation/drug effects , Cytokines/biosynthesis , Hypocreales/chemistry , Killer Cells, Natural/immunology , Male , Mice , Mice, Inbred C57BL , Mycelium/growth & development , Mycelium/metabolism , Spleen/cytology , Spleen/drug effects
17.
J Biosci Bioeng ; 119(1): 19-27, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25027708

ABSTRACT

A fusion construct (Tet-EDIII-Co1) consisting of an M cell-specific peptide ligand (Co1) at the C-terminus of a recombinant tetravalent gene encoding the amino acid sequences of dengue envelope domain III (Tet-EDIII) from four serotypes was expressed and tested for binding activity to the mucosal immune inductive site M cells for the development of an oral vaccine. The yeast episomal expression vector, pYEGPD-TER, which was designed to direct gene expression using the glyceraldehyde-3-phosphate dehydrogenase (GPD) promoter, a functional signal peptide of the amylase 1A protein from rice, and the GAL7 terminator, was used to clone the Tet-EDIII-Co1 gene and resultant plasmids were then used to transform Saccharomyces cerevisiae. PCR and back-transformation into Escherichia coli confirmed the presence of the Tet-EDIII-Co1 gene-containing plasmid in transformants. Northern blot analysis of transformed S. cerevisiae identified the presence of the Tet-EDIII-Co1-specific transcript. Western blot analysis indicated that the produced Tet-EDIII-Co1 protein with the expected molecular weight was successfully secreted into the culture medium. Quantitative Western blot analysis and ELISA revealed that the recombinant Tet-EDIII-Co1 protein comprised approximately 0.1-0.2% of cell-free extracts (CFEs). In addition, 0.1-0.2 mg of Tet-EDIII-Co1 protein per liter of culture filtrate was detected on day 1, and this quantity peaked on day 3 after cultivation. In vivo binding assays showed that the Tet-EDIII-Co1 protein was delivered specifically to M cells in Peyer's patches (PPs) while the Tet-EDIII protein lacking the Co1 ligand did not, which demonstrated the efficient targeting of this antigenic protein through the mucosal-specific ligand.


Subject(s)
Dengue Virus/genetics , Dengue Virus/immunology , Epitopes/genetics , Epitopes/immunology , Peyer's Patches/cytology , Peyer's Patches/immunology , Saccharomyces cerevisiae/genetics , Administration, Oral , Antigens, Viral/analysis , Antigens, Viral/chemistry , Antigens, Viral/genetics , Antigens, Viral/immunology , Dengue Virus/chemistry , Dengue Virus/classification , Epitopes/chemistry , Genetic Vectors/genetics , Immunity, Mucosal/immunology , Ligands , Oryza/enzymology , Oryza/genetics , Plasmids/genetics , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Transformation, Genetic , Viral Vaccines/administration & dosage , Viral Vaccines/chemistry , Viral Vaccines/genetics , Viral Vaccines/immunology
18.
Exp Mol Med ; 46: e85, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24626171

ABSTRACT

Vaccination is one of the most successful applications of immunology and for a long time has depended on parenteral administration protocols. However, recent studies have pointed to the promise of mucosal vaccination because of its ease, economy and efficiency in inducing an immune response not only systemically, but also in the mucosal compartment where many pathogenic infections are initiated. However, successful mucosal vaccination requires the help of an adjuvant for the efficient delivery of vaccine material into the mucosa and the breaking of the tolerogenic environment, especially in oral mucosal immunization. Given that M cells are the main gateway to take up luminal antigens and initiate antigen-specific immune responses, understanding the role and characteristics of M cells is crucial for the development of successful mucosal vaccines. Especially, particular interest has been focused on the regulation of the tolerogenic mucosal microenvironment and the introduction of the luminal antigen into the lymphoid organ by exploiting the molecules of M cells. Here, we review the characteristics of M cells and the immune regulatory factors in mucosa that can be exploited for mucosal vaccine delivery and mucosal immune regulation.


Subject(s)
Antigens, Bacterial/immunology , Antigens, Viral/immunology , Bacterial Vaccines/immunology , Intestinal Mucosa/immunology , Peyer's Patches/immunology , Viral Vaccines/immunology , Administration, Oral , Animals , Bacterial Vaccines/administration & dosage , Humans , Immunity, Mucosal , Intestinal Mucosa/cytology , Peyer's Patches/cytology , Viral Vaccines/administration & dosage
19.
Immune Netw ; 13(4): 157-62, 2013 Aug.
Article in English | MEDLINE | ID: mdl-24009543

ABSTRACT

Application of vaccine materials through oral mucosal route confers great economical advantage in animal farming industry due to much less vaccination cost compared with that of injection-based vaccination. In particular, oral administration of recombinant protein antigen against foot-and-mouth disease virus (FMDV) is an ideal strategy because it is safe from FMDV transmission during vaccine production and can induce antigen-specific immune response in mucosal compartments, where FMDV infection has been initiated, which is hardly achievable through parenteral immunization. Given that effective delivery of vaccine materials into immune inductive sites is prerequisite for effective oral mucosal vaccination, M cell-targeting strategy is crucial in successful vaccination since M cells are main gateway for luminal antigen influx into mucosal lymphoid tissue. Here, we applied previously identified M cell-targeting ligand Co1 to VP1 of FMDV in order to test the possible oral mucosal vaccination against FMDV infection. M cell-targeting ligand Co1-conjugated VP1 interacted efficiently with M cells of Peyer's patch. In addition, oral administration of ligand-conjugated VP1 enhanced the induction of VP1-specific IgG and IgA responses in systemic and mucosal compartments, respectively, in comparison with those from oral administration of VP1 alone. In addition, the enhanced VP1-specific immune response was found to be due to antigen-specific Th2-type cytokine production. Collectively, it is suggested that the M cell-targeting strategy could be applied to develop efficient oral mucosal vaccine against FMDV infection.

20.
Int Immunol ; 25(11): 623-32, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23900425

ABSTRACT

Oral mucosal vaccination is an alternative method to overcome the pitfalls of current injection-based vaccines, such as pain and high cost of vaccination. It is a feasible and economic vaccine application, especially in developing countries. However, achieving effective antigen delivery into mucosal lymphoid organs and efficient immune stimulation are prerequisites to successful oral mucosal vaccination. One promising approach for oral mucosal vaccine development is exploring the potential of M cells via M-cell-targeting ligands that have the potential to deliver ligand-conjugated antigens into mucosal lymphoid organs and evoke conjugated-antigen-specific systemic and mucosal immune responses. Here, we investigated the M-cell-targeting ligand, Co1, in inducing specific immune responses against a pathogenic viral antigen, envelope domain III (EDIII) of dengue virus, to provide the foundation for oral mucosal vaccine development against the pathogen. After oral administration of Co1-conjugated EDIII antigens, we observed efficient antigen delivery into Peyer's patches. We also report the elicitation of EDIII-specific immunity in systemic and mucosal compartments by Co1 ligand (located in the C-terminus of EDIII). Furthermore, the antibodies induced by the ligand-conjugated EDIII antigen showed effective virus-neutralizing activity. The results of this study suggest that the M-cell-targeting strategy using Co1 ligand as a mucosal adjuvant may be applicable for developing oral vaccine candidates against pathogenic viral antigen.


Subject(s)
Antigens, Viral/immunology , Immunity, Mucosal/immunology , Mast Cells/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Administration, Oral , Animals , Antigen-Antibody Reactions , Dengue Virus/immunology , Ligands , Mice , Mice, Inbred BALB C , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL
...