Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Front Immunol ; 11: 360, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32210964

RESUMO

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.


Assuntos
2-Hidroxipropil-beta-Ciclodextrina/farmacologia , Adjuvantes Imunológicos/farmacologia , Vacinas contra Influenza/imunologia , Interleucina-33/fisiologia , 2-Hidroxipropil-beta-Ciclodextrina/administração & dosagem , Administração Intranasal , Animais , Vacinas contra Influenza/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Proteínas Serina-Treonina Quinases/fisiologia , Células Th2/imunologia
3.
Front Immunol ; 10: 2212, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31616416

RESUMO

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.


Assuntos
Aminopiridinas/farmacologia , Asma/tratamento farmacológico , Asma/imunologia , Interleucina-33/imunologia , Nucleotídeos Cíclicos/imunologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/imunologia , Alérgenos/efeitos dos fármacos , Alérgenos/imunologia , Animais , Líquido da Lavagem Broncoalveolar/imunologia , Eosinófilos/efeitos dos fármacos , Eosinófilos/imunologia , Feminino , Imunoglobulina E/imunologia , Imunoglobulina G/imunologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Eosinofilia Pulmonar/tratamento farmacológico , Eosinofilia Pulmonar/imunologia , Pyroglyphidae/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
4.
Arerugi ; 66(6): 809-812, 2017.
Artigo em Japonês | MEDLINE | ID: mdl-28701647

RESUMO

We report a case of jellyfish allergy diagnosed via an oral food challenge. A 14-year-old boy had no history of jellyfish stings and had been eating commercially available jellyfish products twice yearly for the past 5-6 years. Five minutes after eating a commercially available boiled jellyfish product (100g), he experienced nausea, wheezing, and erythema and had visited our hospital. We suspected an anaphylactic reaction and treated him with intramuscular adrenaline injection, corticosteroid and antihistamine infusions, volume resuscitation, and salbutamol sulfate inhalation, which resulted in an improvement of the symptoms. One-month later in our hospital, we administered an oral food challenge of the same boiled jellyfish product bought at the same grocery store to the patient. After ingesting 14g of boiled jellyfish, he experienced erythema, wheezing, nausea, and abdominal pain. Several reports have described anaphylaxis caused by the ingestion of jellyfish, but the allergens in jellyfish have not been analyzed. A skin prick test for poly-gamma-glutamic acid (PGA) which is a component of jellyfish stings was negative. This suggests that he was sensitized to some allergen other than PGA via a route different from that of jellyfish sting. Our skin prick test for several kinds of edible jellyfish suggests that allergenicity may be different for different jellyfish.


Assuntos
Anafilaxia/imunologia , Hipersensibilidade Alimentar/complicações , Hipersensibilidade Alimentar/imunologia , Cifozoários/imunologia , Adolescente , Alérgenos/imunologia , Animais , Ingestão de Alimentos , Humanos , Masculino
5.
J Immunol ; 198(12): 4707-4715, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28507029

RESUMO

DNA vaccines are attractive immunogens for priming humoral and cellular immune responses to the encoded Ag. However, their ability to induce Ag-specific CD8+ T cell responses requires improvement. Among the strategies for improving DNA vaccine immunogenicity are booster vaccinations, alternate vaccine formulations, electroporation, and genetic adjuvants, but few, such as extracellular vesicles (EVs), target natural Ag delivery systems. By focusing on CD63, a tetraspanin protein expressed on various cellular membranes, including EVs, we examined whether a DNA vaccine encoding an Ag fused to CD63 delivered into EVs would improve vaccine immunogenicity. In vitro transfection with plasmid DNA encoding an OVA Ag fused to CD63 (pCD63-OVA) produced OVA-carrying EVs. Immunizations with the purified OVA-carrying EVs primed naive mice to induce OVA-specific CD4+ and CD8+ T cells, whereas immunization with EVs purified from cells transfected with control plasmids encoding OVA protein alone or a calnexin-OVA fusion protein delivered into the endoplasmic reticulum failed to do so. Vaccinating mice with pCD63-OVA induced potent Ag-specific T cell responses, particularly those from CD8+ T cells. CD63 delivery into EVs led to better CD8+ T cell responses than calnexin delivery into the endoplasmic reticulum. When we used a mouse tumor implantation model to evaluate pCD63-OVA as a therapeutic vaccine, the EV-delivered DNA vaccination significantly inhibited tumor growth compared with the control DNA vaccinations. These results indicate that EV Ag delivery via DNA vaccination offers a new strategy for eliciting strong CD8+ T cell responses to the encoded Ag, making it a potentially useful cancer vaccine.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vesículas Extracelulares/imunologia , Ativação Linfocitária , Tetraspanina 30/imunologia , Vacinas de DNA/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Apresentação de Antígeno/imunologia , Linfócitos T CD4-Positivos/imunologia , Vacinas Anticâncer/imunologia , Feminino , Imunidade Celular , Imunização Secundária , Imunogenicidade da Vacina , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Tetraspanina 30/genética , Vacinas de DNA/administração & dosagem
6.
Immunity ; 45(6): 1299-1310, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-28002730

RESUMO

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.


Assuntos
Brônquios/imunologia , Interleucina-1alfa/imunologia , Tecido Linfoide/imunologia , Macrófagos Alveolares/patologia , Material Particulado/toxicidade , Compostos de Alumínio/toxicidade , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Dióxido de Silício/toxicidade
7.
Sci Rep ; 6: 29165, 2016 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-27374884

RESUMO

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.


Assuntos
Adjuvantes Imunológicos/farmacologia , Lipídeos/química , Mucosa/efeitos dos fármacos , RNA/farmacologia , Células A549 , Alarminas/metabolismo , Animais , Formação de Anticorpos/efeitos dos fármacos , Feminino , Humanos , Inflamassomos/metabolismo , Lipídeos/farmacologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mucosa/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Ácidos Nucleicos/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo
8.
Oncotarget ; 7(31): 48860-48869, 2016 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-27384490

RESUMO

Recent evidence suggest that a ß-glucan derived from mushroom Schizophyllan(SPG) complexed with a humanized TLR9 agonistic CpG DNA, K3 (K3-SPG) is a promising vaccine adjuvant that induces robust CD8 T cell responses to co-administered antigen. However, it has not been investigated whether K3-SPG alone can act as an anti-cancer immunotherapeutic agent or not. Here, we demonstrate that intravenous injection of K3-SPG, but not CpG alone, is accumulated in the tumor microenvironment and triggered immunogenic cell death (ICD) of tumor cells by local induction of type-I interferon (IFN) as well as IL-12. Resultant innate immune activation as well as subsequent tumor-specific CD8 T cell responses were contributed the tumor growth suppression. This anti-tumor effect of K3-SPG monotherapy was also confirmed by using various tumor models including pancreatic cancer peritoneal dissemination model. Taken together, nano-particulate TLR9 agonist injected intravenously can scout out tumor microenvironment to provoke local innate immune activation and release dead tumor cells into circulation that may induce broader and protective tumor antigen-specific CD8 T cells.


Assuntos
Nanopartículas/química , Neoplasias/imunologia , Neoplasias/terapia , Sizofirano/farmacologia , Receptor Toll-Like 9/agonistas , Microambiente Tumoral/efeitos dos fármacos , Adjuvantes Imunológicos/farmacologia , Animais , Formação de Anticorpos , Linfócitos T CD8-Positivos/imunologia , Ilhas de CpG/efeitos dos fármacos , Citocinas/farmacologia , Humanos , Imunidade Inata , Interferon Tipo I/farmacologia , Interferon gama/imunologia , Interleucina-12/imunologia , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias/sangue , Oligodesoxirribonucleotídeos/administração & dosagem , Fagocitose
9.
Vaccine ; 34(27): 3191-3198, 2016 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-27160037

RESUMO

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.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , beta-Ciclodextrinas/administração & dosagem , 2-Hidroxipropil-beta-Ciclodextrina , Administração Intranasal , Animais , Anticorpos Antivirais/sangue , Sistemas de Liberação de Medicamentos , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Imunidade nas Mucosas , Vacinas contra Influenza/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia
10.
Eur J Immunol ; 46(5): 1142-51, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26786557

RESUMO

CpG oligodeoxynucleotide (ODN) is one of promising nucleic acid-based adjuvants. We recently improved its ability to enhance CD8(+) T-cell responses to coadministered protein antigen without conjugation or emulsion, by forming a nanoparticulate complex between CpG ODN (K3) and mushroom-derived ß-glucan schizophyllan (SPG), namely K3-SPG. Here, we sought to elucidate the cellular immunological mechanisms by which K3-SPG induce such potent CD8(+) T-cell responses to coadministered antigen. By focusing on two DC subsets, plasmacytoid DCs and CD8α(+) DCs, as well as the secreted cytokines, IFN-α and IL-12, we found that K3-SPG strongly activates mouse plasmacytoid DCs to secrete IFN-α and CD8α(+) DCs to secrete IL-12, respectively. Although a single cytokine deficiency had no impact on adjuvant effects, the lack of both type I IFN and IL-12 in mice resulted in a significant reduction of Th1 type immune responses and CD8(+) T-cell responses elicited by protein vaccine model. By sharp contrast, type I IFN, but not IL-12, was required for the production of IFN-γ by human PBMCs as well as antigen-specific CD8(+) T-cell proliferation. Taken together, K3-SPG may overcome the species barrier for CpG ODN to enhance antigen-specific CD8(+) T-cell responses despite the differential role of IL-12 between human and mice.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Interferon-alfa/imunologia , Interleucina-12/imunologia , Oligodesoxirribonucleotídeos/imunologia , beta-Glucanas/imunologia , Adjuvantes Imunológicos , Adulto , Animais , Proliferação de Células , Humanos , Interferon gama/imunologia , Interleucina-12/deficiência , Interleucina-12/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Ativação Linfocitária , Masculino , Camundongos , Oligodesoxirribonucleotídeos/química , Oligodesoxirribonucleotídeos/farmacologia , Células Th1/imunologia , Receptor Toll-Like 9/agonistas
11.
J Immunol ; 194(6): 2673-82, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25681338

RESUMO

Cyclodextrins are commonly used as a safe excipient to enhance the solubility and bioavailability of hydrophobic pharmaceutical agents. Their efficacies and mechanisms as drug-delivery systems have been investigated for decades, but their immunological properties have not been examined. In this study, we reprofiled hydroxypropyl-ß-cyclodextrin (HP-ß-CD) as a vaccine adjuvant and found that it acts as a potent and unique adjuvant. HP-ß-CD triggered the innate immune response at the injection site, was trapped by MARCO(+) macrophages, increased Ag uptake by dendritic cells, and facilitated the generation of T follicular helper cells in the draining lymph nodes. It significantly enhanced Ag-specific Th2 and IgG Ab responses as potently as did the conventional adjuvant, aluminum salt (alum), whereas its ability to induce Ag-specific IgE was less than that of alum. At the injection site, HP-ß-CD induced the temporary release of host dsDNA, a damage-associated molecular pattern. DNase-treated mice, MyD88-deficient mice, and TBK1-deficient mice showed significantly reduced Ab responses after immunization with this adjuvant. Finally, we demonstrated that HP-ß-CD-adjuvanted influenza hemagglutinin split vaccine protected against a lethal challenge with a clinically isolated pandemic H1N1 influenza virus, and the adjuvant effect of HP-ß-CD was demonstrated in cynomolgus macaques. Our results suggest that HP-ß-CD acts as a potent MyD88- and TBK1-dependent T follicular helper cell adjuvant and is readily applicable to various vaccines.


Assuntos
Antígenos/imunologia , Inflamação/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Células Th2/imunologia , beta-Ciclodextrinas/imunologia , 2-Hidroxipropil-beta-Ciclodextrina , Adjuvantes Imunológicos/administração & dosagem , Animais , Formação de Anticorpos/efeitos dos fármacos , Formação de Anticorpos/genética , Formação de Anticorpos/imunologia , Antígenos/administração & dosagem , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/imunologia , Inflamação/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Linfonodos/metabolismo , Macaca fascicularis , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência por Excitação Multifotônica , Análise de Sequência com Séries de Oligonucleotídeos , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia , Baço/efeitos dos fármacos , Baço/imunologia , Baço/metabolismo , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/metabolismo , Células Th2/efeitos dos fármacos , Células Th2/metabolismo , Transcriptoma/efeitos dos fármacos , Transcriptoma/imunologia , beta-Ciclodextrinas/administração & dosagem
12.
Eur J Immunol ; 45(4): 1159-69, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25529558

RESUMO

Agonists for TLR9 and Stimulator of IFN Gene (STING) act as vaccine adjuvants that induce type-1 immune responses. However, currently available CpG oligodeoxynucleotide (ODN) (K-type) induces IFNs only weakly and STING ligands rather induce type-2 immune responses, limiting their potential therapeutic applications. Here, we show a potent synergism between TLR9 and STING agonists. Together, they make an effective type-1 adjuvant and an anticancer agent. The synergistic effect between CpG ODN (K3) and STING-ligand cyclic GMP-AMP (cGAMP), culminating in NK cell IFN-γ (type-II IFN) production, is due to the concurrent effects of IL-12 and type-I IFNs, which are differentially regulated by IRF3/7, STING, and MyD88. The combination of CpG ODN with cGAMP is a potent type-1 adjuvant, capable of inducing strong Th 1-type responses, as demonstrated by enhanced antigen-specific IgG2c and IFN-γ production, as well as cytotoxic CD8(+) T-cell responses. In our murine tumor models, intratumoral injection of CpG ODN and cGAMP together reduced tumor size significantly compared with the singular treatments, acting as an antigen-free anticancer agent. Thus, the combination of CpG ODN and a STING ligand may offer therapeutic application as a potent type-II IFN inducer.


Assuntos
Interferon gama/biossíntese , Proteínas de Membrana/agonistas , Neoplasias/terapia , Nucleotídeos Cíclicos/farmacologia , Oligodesoxirribonucleotídeos/farmacologia , Linfócitos T Citotóxicos/imunologia , Receptor Toll-Like 9/agonistas , Adjuvantes Imunológicos/farmacologia , Animais , Linhagem Celular Tumoral , Sinergismo Farmacológico , Feminino , Imunoglobulina G/biossíntese , Imunoglobulina G/imunologia , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/metabolismo , Interferon Tipo I/metabolismo , Interferon gama/imunologia , Interleucina-12/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...