Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Int Immunol ; 34(7): 353-364, 2022 07 04.
Article in English | MEDLINE | ID: mdl-35419609

ABSTRACT

Agonists for TLR9 and stimulator of IFN genes (STING) offer therapeutic applications as both anti-tumor agents and vaccine adjuvants, though their clinical applications are limited; the clinically available TLR9 agonist is a weak IFN inducer and STING agonists induce undesired type 2 immunity. Yet, combining TLR9 and STING agonists overcame these limitations by synergistically inducing innate and adaptive IFNγ to become an advantageous type 1 adjuvant, suppressing type 2 immunity, in addition to exerting robust anti-tumor activities when used as a monotherapeutic agent for cancer immunotherapy. Here, we sought to decipher the immunological mechanisms behind the synergism mediated by TLR9 and STING agonists and found that their potent anti-tumor immunity in a Pan02 peritoneal dissemination model of pancreatic cancer was achieved only when agonists for TLR9 and STING were administered locally, and was via mechanisms involving CD4 and CD8 T cells as well as the co-operative action of IL-12 and type I IFNs. Rechallenge studies of long-term cancer survivors suggested that the elicitation of Pan02-specific memory responses provides protection against the secondary tumor challenge. Mechanistically, we found that TLR9 and STING agonists synergistically induce IL-12 and type I IFN production in murine APCs. The synergistic effect of the TLR9 and STING agonists on IL-12p40 was at protein, mRNA and promoter activation levels, and transcriptional regulation was mediated by a 200 bp region situated 983 bp upstream of the IL-12p40 transcription initiation site. Such intracellular transcriptional synergy may hold a key in successful cancer immunotherapy and provide further insights into dual agonism of innate immune sensors during host homeostasis and diseases.


Subject(s)
Membrane Proteins , Neoplasms , Toll-Like Receptor 9 , Adjuvants, Immunologic/pharmacology , Animals , Immunotherapy , Interleukin-12 , Interleukin-12 Subunit p40 , Membrane Proteins/metabolism , Mice , Toll-Like Receptor 9/metabolism
2.
Front Immunol ; 11: 360, 2020.
Article in English | MEDLINE | ID: mdl-32210964

ABSTRACT

Vaccine adjuvants are traditionally used to augment and modulate the immunogenicity of vaccines, although in many cases it is unclear which specific molecules contribute to their stimulatory activity. We previously reported that both subcutaneous and intranasal administration of hydroxypropyl-ß-cyclodextrin (HP-ß-CD), a pharmaceutical excipient widely used to improve solubility, can act as an effective adjuvant for an influenza vaccine. However, the mechanisms by which mucosal immune pathway is critical for the intranasal adjuvant activity of HP-ß-CD have not been fully delineated. Here, we show that intranasally administered HP-ß-CD elicits a temporary release of IL-33 from alveolar epithelial type 2 cells in the lung; notably, IL-33 expression in these cells is not stimulated following the use of other vaccine adjuvants. The experiments using gene deficient mice suggested that IL-33/ST2 signaling is solely responsible for the adjuvant effect of HP-ß-CD when it is administered intranasally. In contrast, the subcutaneous injection of HP-ß-CD and the intranasal administration of alum, as a damage-associated molecular patterns (DAMPs)-inducing adjuvant, or cholera toxin, as a mucosal adjuvant, enhanced humoral immunity in an IL-33-independent manner, suggesting that the IL-33/ST2 pathway is unique to the adjuvanticity of intranasally administered HP-ß-CD. Furthermore, the release of IL-33 was involved in the protective immunity against influenza virus infection which is induced by the intranasal administration of HP-ß-CD-adjuvanted influenza split vaccine. In conclusion, our results suggest that an understanding of administration route- and tissue-specific immune responses is crucial for the design of unique vaccine adjuvants.


Subject(s)
2-Hydroxypropyl-beta-cyclodextrin/pharmacology , Adjuvants, Immunologic/pharmacology , Influenza Vaccines/immunology , Interleukin-33/physiology , 2-Hydroxypropyl-beta-cyclodextrin/administration & dosage , Administration, Intranasal , Animals , Influenza Vaccines/administration & dosage , Mice , Mice, Inbred C57BL , Organ Specificity , Protein Serine-Threonine Kinases/physiology , Th2 Cells/immunology
3.
Front Immunol ; 10: 2212, 2019.
Article in English | MEDLINE | ID: mdl-31616416

ABSTRACT

Extracellular host-derived DNA, as one of damage associated molecular patterns (DAMPs), is associated with allergic type 2 immune responses. Immune recognition of such DNA generates the second messenger cyclic GMP-AMP (cGAMP) and induces type-2 immune responses; however, its role in allergic diseases, such as asthma, has not been fully elucidated. This study aimed to determine whether cGAMP could induce asthma when used as an adjuvant. We intranasally sensitized mice with cGAMP together with house dust mite antigen (HDM), followed by airway challenge with HDM. We then assessed the levels of eosinophils in the broncho-alveolar lavage fluid (BALF) and serum HDM-specific antibodies. cGAMP promoted HDM specific allergic asthma, characterized by significantly increased HDM specific IgG1 and total IgE in the serum and infiltration of eosinophils in the BALF. cGAMP stimulated lung fibroblast cells to produce IL-33 in vitro, and mice deficient for IL-33 or IL-33 receptor (ST2) failed to develop asthma enhancement by cGAMP. Not only Il-33-/- mice, but also Sting-/-, Tbk1-/-, and Irf3-/-Irf7-/- mice which lack the cGAMP-mediated innate immune activation failed to increase eosinophils in the BALF than that from wild type mice. Consistently, intranasal and oral administration of amlexanox, a TBK1 inhibitor, decreased cGAMP-induced lung allergic inflammation. Thus, cGAMP functions as a type 2 adjuvant in the lung and can promote allergic asthma in manners that dependent on the intracellular STING/TBK1/IRF3/7 signaling pathway and the resultant intercellular signaling pathway via IL-33 and ST2 might be a novel therapeutic target for allergic asthma.


Subject(s)
Aminopyridines/pharmacology , Asthma/drug therapy , Asthma/immunology , Interleukin-33/immunology , Nucleotides, Cyclic/immunology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/immunology , Allergens/drug effects , Allergens/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Eosinophils/drug effects , Eosinophils/immunology , Female , Immunoglobulin E/immunology , Immunoglobulin G/immunology , Lung/drug effects , Lung/immunology , Mice , Mice, Inbred C57BL , Pulmonary Eosinophilia/drug therapy , Pulmonary Eosinophilia/immunology , Pyroglyphidae/immunology , Signal Transduction/drug effects , Signal Transduction/immunology
4.
Mucosal Immunol ; 12(5): 1082-1091, 2019 09.
Article in English | MEDLINE | ID: mdl-31142830

ABSTRACT

Leukotriene B4 receptor 1 (BLT1) triggers the migration of granulocytes and activated T cells; however, its role in B-cell function remains unclear. Here we report that BLT1 is required to induce the production of antigen-specific IgA against oral vaccine through mediating innate immune signals from commensal bacteria. B cells acquire BLT1 expression during their differentiation to IgA+ B cells and plasma cells in Peyer's patches and the small intestinal lamina propria, respectively. BLT1 KO mice exhibited impaired production of antigen-specific fecal IgA to oral vaccine despite normal IgG responses to systemically immunized antigen. Expression of MyD88 was decreased in BLT1 KO gut B cells and consequently led to diminished proliferation of commensal bacteria-dependent plasma cells. These results indicate that BLT1 enhances the proliferation of commensal bacteria-dependent IgA+ plasma cells through the induction of MyD88 and thereby plays a key role in the production of antigen-specific intestinal IgA.


Subject(s)
Epitopes/immunology , Gastrointestinal Microbiome/immunology , Immunity, Innate , Immunoglobulin A, Secretory/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Receptors, Leukotriene B4/genetics , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Immunization , Intestinal Mucosa/microbiology , Male , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Peyer's Patches/immunology , Peyer's Patches/metabolism , Plasma Cells/immunology , Plasma Cells/metabolism , Receptors, Leukotriene B4/metabolism , Signal Transduction , Vaccines/administration & dosage , Vaccines/immunology
5.
Front Immunol ; 9: 2619, 2018.
Article in English | MEDLINE | ID: mdl-30515151

ABSTRACT

Recently, it was reported that 2-hydroxypropyl-ß-cyclodextrin (HP-ß-CyD), a common pharmaceutical additive, can act as a vaccine adjuvant to enhance protective type-2 immunogenicity to co-administered seasonal influenza split vaccine by inducing host-derived damage-associated molecular patterns (DAMPs). However, like most other DAMP-inducing adjuvants such as aluminum hydroxide (Alum), HP-ß-CyD may not be sufficient for the induction of protective type-1 (cellular) immune responses, thereby leaving room for improvement. Here, we demonstrate that a combination of HP-ß-CyD with a humanized TLR9 agonist, K3 CpG-ODN, a potent pathogen-associated molecular pattern (PAMP), enhanced the protective efficacy of the co-administered influenza split vaccine by inducing antigen-specific type-2 and type-1 immune responses, respectively. Moreover, substantial antigen-specific IgE induction by HP-ß-CyD, which can cause an allergic response to immunized antigen was completely suppressed by the addition of K3 CpG-ODN. Furthermore, HP-ß-CyD- and K3 CpG-ODN-adjuvanted influenza split vaccination protected the mice against lethal challenge with high doses of heterologous influenza virus, which could not be protected against by single adjuvant vaccines. Further experiments using gene deficient mice revealed the unique immunological mechanism of action in vivo, where type-2 and type-1 immune responses enhanced by the combined adjuvants were dependent on TBK1 and TLR9, respectively, indicating their parallel signaling pathways. Finally, the analysis of immune responses in the draining lymph node suggested that HP-ß-CyD promotes the uptake of K3 CpG-ODN by plasmacytoid dendritic cells and B cells, which may contributes to the activation of these cells and enhanced production of IgG2c. Taken together, the results above may offer potential clinical applications for the combination of DAMP-inducing adjuvant and PAMP adjuvant to improve vaccine immunogenicity and efficacy by enhancing both type-2 and type-1 immune responses in a parallel manner.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Influenza A Virus, H1N1 Subtype/physiology , Influenza Vaccines/immunology , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , Th1 Cells/immunology , Th2 Cells/immunology , 2-Hydroxypropyl-beta-cyclodextrin/immunology , Adjuvants, Immunologic , Alarmins/metabolism , Animals , Antibodies, Viral/blood , Cells, Cultured , Female , Humans , Immunogenicity, Vaccine , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligodeoxyribonucleotides/immunology , Pathogen-Associated Molecular Pattern Molecules/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/genetics , Vaccination
6.
Immunity ; 45(6): 1299-1310, 2016 12 20.
Article in English | MEDLINE | ID: mdl-28002730

ABSTRACT

Particulate pollution is thought to function as an adjuvant that can induce allergic responses. However, the exact cell types and immunological factors that initiate the lung-specific immune responses are unclear. We found that upon intratracheal instillation, particulates such as aluminum salts and silica killed alveolar macrophages (AMs), which then released interleukin-1α (IL-1α) and caused inducible bronchus-associated lymphoid tissue (iBALT) formation in the lung. IL-1α release continued for up to 2 weeks after particulate exposure, and type-2 allergic immune responses were induced by the inhalation of antigen during IL-1α release and iBALT formation, even long after particulate instillation. Recombinant IL-1α was sufficient to induce iBALTs, which coincided with subsequent immunoglobulin E responses, and IL-1-receptor-deficient mice failed to induce iBALT formation. Therefore, the AM-IL-1α-iBALT axis might be a therapeutic target for particulate-induced allergic inflammation.


Subject(s)
Bronchi/immunology , Interleukin-1alpha/immunology , Lymphoid Tissue/immunology , Macrophages, Alveolar/pathology , Particulate Matter/toxicity , Aluminum Compounds/toxicity , Animals , Female , Mice , Mice, Inbred C57BL , Silicon Dioxide/toxicity
7.
Sci Rep ; 6: 29165, 2016 07 04.
Article in English | MEDLINE | ID: mdl-27374884

ABSTRACT

Nasal vaccination has the potential to elicit systemic and mucosal immunity against pathogens. However, split and subunit vaccines lack potency at stimulating mucosal immunity, and an adjuvant is indispensable for eliciting potent mucosal immune response to nasal vaccines. Endocine, a lipid-based mucosal adjuvant, potentiates both systemic and mucosal immune responses. Although Endocine has shown efficacy and tolerability in animal and clinical studies, its mechanism of action remains unknown. It has been reported recently that endogenous danger signals are essential for the effects of some adjuvants such as alum or MF59. However, the contribution of danger signals to the adjuvanticity of Endocine has not been explored. Here, we show that RNA is likely to be an important mediator for the adjuvanticity of Endocine. Administration of Endocine generated nucleic acids release, and activated dendritic cells (DCs) in draining lymph nodes in vivo. These results suggest the possibility that Endocine indirectly activates DCs via damage-associated molecular patterns. Moreover, the adjuvanticity of Endocine disappeared in mice lacking TANK-binding kinase 1 (Tbk1), which is a downstream molecule of nucleic acid sensing signal pathway. Furthermore, co-administration of RNase A reduced the adjuvanticity of Endocine. These data suggest that RNA is important for the adjuvanticity of Endocine.


Subject(s)
Adjuvants, Immunologic/pharmacology , Lipids/chemistry , Mucous Membrane/drug effects , RNA/pharmacology , A549 Cells , Alarmins/metabolism , Animals , Antibody Formation/drug effects , Female , Humans , Inflammasomes/metabolism , Lipids/pharmacology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mucous Membrane/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nucleic Acids/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
8.
Vaccine ; 34(27): 3191-3198, 2016 06 08.
Article in English | MEDLINE | ID: mdl-27160037

ABSTRACT

Intranasal vaccination with inactivated influenza viral antigens is an attractive and valid alternative to currently available influenza (flu) vaccines; many of which seem to need efficient and safe adjuvant, however. In this study, we examined whether hydroxypropyl-ß-cyclodextrin (HP-ß-CD), a widely used pharmaceutical excipient to improve solubility and drug delivery, can act as a mucosal adjuvant for intranasal flu vaccines. We found that intranasal immunization of mice with hemagglutinin split- as well as inactivated whole-virion influenza vaccine with HP-ß-CD resulted in secretion of antigen-specific IgA and IgGs in the airway mucosa and the serum as well. As a result, both HP-ß-CD adjuvanted-flu intranasal vaccine protected mice against lethal challenge with influenza virus, equivalent to those induced by experimental cholera toxin-adjuvanted ones. Of note, intranasal use of HP-ß-CD as an adjuvant induced significantly lower antigen-specific IgE responses than that induced by aluminum salt adjuvant. These results suggest that HP-ß-CD may be a potent mucosal adjuvant for seasonal and pandemic influenza vaccine.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , beta-Cyclodextrins/administration & dosage , 2-Hydroxypropyl-beta-cyclodextrin , Administration, Intranasal , Animals , Antibodies, Viral/blood , Drug Delivery Systems , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunity, Mucosal , Influenza Vaccines/administration & dosage , Mice , Mice, Inbred C57BL , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...