Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Front Immunol ; 11: 564497, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33162977

RESUMO

The mechanisms of trained immunity have been extensively described in vitro and the beneficial effects are starting to be deciphered in in vivo settings. Prototypical compounds inducing trained immunity, such as ß-glucans, act through epigenetic reprogramming and metabolic changes of innate immune cells. The recent advances in this field have opened new areas for the development of Trained immunity-based adjuvants (TIbAs). In this study, we assessed in dogs the potential immune training effects of ß-glucans as well as their capacity to enhance the adaptive immune response of an inactivated rabies vaccine (Rabisin®). Injection of ß-glucan from Euglena gracilis was performed 1 month before vaccination with Rabisin® supplemented or not with the same ß-glucan used as adjuvant. Trained innate immunity parameters were assessed during the first month of the trial. The second phase of the study was focused on the ability of ß-glucan to enhance adaptive immune responses measured by multiple immunological parameters. B and T-cell specific responses were monitored to evaluate the immunogenicity of the rabies vaccine adjuvanted with ß-glucan or not. Our preliminary results support that adjuvantation of Rabisin® vaccine with ß-glucan elicit a higher B-lymphocyte immune response, the prevailing factor of protection against rabies. ß-glucan also tend to stimulate the T cell response as shown by the cytokine secretion profile of PBMCs re-stimulated ex vivo. Our data are providing new insights on the impact of trained immunity on the adaptive immune response to vaccines in dogs. The administration of ß-glucan, 1 month before or simultaneously to Rabisin® vaccination give promising results for the generation of new TIbA candidates and their potential to provide increased immunogenicity of specific vaccines.


Assuntos
Adjuvantes Imunológicos/farmacologia , Imunogenicidade da Vacina/efeitos dos fármacos , Vacina Antirrábica/imunologia , Raiva/prevenção & controle , Raiva/veterinária , Vacinação/métodos , Vacinação/veterinária , beta-Glucanas/farmacologia , Imunidade Adaptativa/efeitos dos fármacos , Animais , Linfócitos B/imunologia , Citocinas/metabolismo , Cães , Euglena gracilis/química , Feminino , Imunidade Inata/efeitos dos fármacos , Masculino , Distribuição Aleatória , Linfócitos T/imunologia , Resultado do Tratamento , Vacinas de Produtos Inativados/imunologia
2.
Front Immunol ; 11: 566893, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33162983

RESUMO

Several observations in the world of comparative immunology in plants, insects, fish and eventually mammals lead to the discovery of trained immunity in the early 2010's. The first demonstrations provided evidence that innate immune cells were capable of developing memory after a first encounter with some pathogens. Trained immunity in mammals was initially described in monocytes with the Bacille Calmette-Guerin vaccine (BCG) or prototypical agonists like ß-glucans. This phenomenon relies on epigenetic and metabolic modifications leading to an enhanced secretion of inflammatory cytokines when the host encounters homologous or heterologous pathogens. The objective of our research was to investigate the trained immunity, well-described in mouse and human, in other species of veterinary importance. For this purpose, we adapted an in vitro model of trained innate immunity in dogs. Blood enriched monocytes were stimulated with ß-glucans and we confirmed that it induced an increased production of pro-inflammatory and anti-microbial compounds in response to bacterial stimuli. These results constitute the first demonstration of trained immunity in dogs and confirm its signatures in other mammalian species, with an implication of cellular mechanisms similar to those described in mice and humans regarding cellular epigenetics and metabolic regulations.


Assuntos
Imunidade Inata/imunologia , Monócitos/efeitos dos fármacos , beta-Glucanas/farmacologia , Animais , Células Cultivadas , Citocinas/imunologia , Cães , Feminino , Fatores Imunológicos/farmacologia , Masculino , Monócitos/imunologia , Fagocitose/efeitos dos fármacos
3.
Front Immunol ; 9: 2869, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30564249

RESUMO

Epidemiological studies regarding many successful vaccines suggest that vaccination may lead to a reduction in child mortality and morbidity worldwide, on a grander scale than is attributable to protection against the specific target diseases of these vaccines. These non-specific effects (NSEs) of the Bacille Calmette-Guérin (BCG) vaccine, for instance, implicate adaptive and innate immune mechanisms, with recent evidence suggesting that trained immunity might be a key instrument at play. Collectively referring to the memory-like characteristics of innate immune cells, trained immunity stems from epigenetic reprogramming that these innate immune cells undergo following exposure to a primary stimulus like BCG. The epigenetic changes subsequently regulate cytokine production and cell metabolism and in turn, epigenetic changes are regulated by these effects. Novel -omics technologies, combined with in vitro models for trained immunity and other immunological techniques, identify the biological pathways within innate cells that enable training by BCG. Future research should aim to identify biomarkers for vaccine heterologous effects, such that they can be applied to epidemiological studies. Linking biological mechanisms to the reduction in all-cause mortality observed in epidemiological studies will strengthen the evidence in favor of vaccine NSEs. The universal acceptance of these NSEs would demand a re-evaluation of current vaccination policies, such as the childhood vaccination recommendations by the World Health Organization, in order to produce the maximum impact on childhood mortality.


Assuntos
Vacina BCG/imunologia , Imunidade Heteróloga , Memória Imunológica , Mycobacterium bovis/imunologia , Tuberculose/prevenção & controle , Vacina BCG/administração & dosagem , Epigênese Genética/imunologia , Humanos , Imunidade Celular , Imunidade Inata , Vacinação/métodos , Vacinação/normas , Organização Mundial da Saúde
4.
PLoS One ; 11(8): e0162109, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27574993

RESUMO

Sepsis is characterized by pro- and anti-inflammatory responses following infection. While inflammation is responsible for widespread organ damage, anti-inflammatory mediators lead to immunoparalysis increasing susceptibility to secondary infections (nosocomial pneumonia). We aimed to investigate the impact of bacterial load on survival and cytokine release in a two-hit murine (C57BL/6J) model of CLP followed by P. aeruginosa pneumonia. Plasmatic TNFα, IL-6, IL-10, sTNFr I and II were quantified until 13 days. At D5, splenocytes were processed for immunological assays or mice were intratracheally instilled with Pseudomonas aeruginosa (5.106, 2.107 and 108 CFU) to evaluate survival and cytokines production. TNFα, sTNFrs, IL-6 and IL-10 increased 2h post CLP. TNFα and sTNFrs declined respectively one and two days later. In CLP mice, IL-6 and IL-10 remained high for the whole experiment, as compared to Sham. At D5, for CLP mice, whereas total T cells population (CD3+) decreased, Treg fraction (CD4+/CD25+) increased. In parallel, T cells proliferation and LPS-stimulated splenocytes ability to release TNFα decreased. At D13, survival was 100% after 5.106 CFU, 50% for CLP mice after 2.107 CFU and 0% for CLP and Sham after 108 CFU. After instillation, IL-10 and IL-6 increased and appeared to be dose and time dependent. Pseudomonas was detected in all CLP and Sham's lungs; in spleen and liver only in CLP at 2.107 CFU, and in CLP and Sham at 108 CFU. We demonstrated that post-CLP immunosuppression followed by Pseudomonas aeruginosa lung instillation increases mortality reactivates cytokines secretion and is associated with systemic dissemination in septic mice depending on bacterial load.


Assuntos
Citocinas/sangue , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/crescimento & desenvolvimento , Sepse/complicações , Animais , Carga Bacteriana , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Sepse/imunologia , Baço/citologia , Baço/imunologia , Análise de Sobrevida , Linfócitos T/citologia , Linfócitos T/imunologia
5.
Sci Transl Med ; 5(172): 172ra20, 2013 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-23408053

RESUMO

Although many human cancers are located in mucosal sites, most cancer vaccines are tested against subcutaneous tumors in preclinical models. We therefore wondered whether mucosa-specific homing instructions to the immune system might influence mucosal tumor outgrowth. We showed that the growth of orthotopic head and neck or lung cancers was inhibited when a cancer vaccine was delivered by the intranasal mucosal route but not the intramuscular route. This antitumor effect was dependent on CD8⁺ T cells. Indeed, only intranasal vaccination elicited mucosal-specific CD8⁺ T cells expressing the mucosal integrin CD49a. Blockade of CD49a decreased intratumoral CD8⁺ T cell infiltration and the efficacy of cancer vaccine on mucosal tumor. We then showed that after intranasal vaccination, dendritic cells from lung parenchyma, but not those from spleen, induced the expression of CD49a on cocultured specific CD8⁺ T cells. Tumor-infiltrating lymphocytes from human mucosal lung cancer also expressed CD49a, which supports the relevance and possible extrapolation of these results in humans. We thus identified a link between the route of vaccination and the induction of a mucosal homing program on induced CD8⁺ T cells that controlled their trafficking. Immunization route directly affected the efficacy of the cancer vaccine to control mucosal tumors.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Quimiotaxia de Leucócito , Neoplasias de Cabeça e Pescoço/terapia , Imunidade nas Mucosas , Neoplasias Pulmonares/terapia , Mucosa Nasal/imunologia , Vacinas contra Papillomavirus/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Administração Intranasal , Animais , Antígenos CD/metabolismo , Vacinas Anticâncer/imunologia , Proliferação de Células , Células Cultivadas , Células Dendríticas/imunologia , Feminino , Neoplasias de Cabeça e Pescoço/imunologia , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Injeções Intramusculares , Cadeias alfa de Integrinas/metabolismo , Integrina alfa1/metabolismo , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Linfonodos/imunologia , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Vacinas contra Papillomavirus/imunologia , Toxinas Shiga/administração & dosagem , Baço/imunologia , Carga Tumoral
6.
J Immunol ; 179(5): 3371-9, 2007 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-17709554

RESUMO

The nontoxic B subunit of Shiga toxin (STxB) targets in vivo Ag to dendritic cells that preferentially express the glycolipid Gb(3) receptor. After administration of STxB chemically coupled to OVA (STxB-OVA) or E7, a polypeptide derived from HPV, in mice, we showed that the addition of alpha-galactosylceramide (alpha-GalCer) resulted in a dramatic improvement of the STxB Ag delivery system, as reflected by the more powerful and longer lasting CD8(+) T cell response observed even at very low dose of immunogen (50 ng). This synergy was not found with other adjuvants (CpG, poly(I:C), IFN-alpha) also known to promote dendritic cell maturation. With respect to the possible mechanism explaining this synergy, mice immunized with alpha-GalCer presented in vivo the OVA(257-264)/K(b) complex more significantly and for longer period than mice vaccinated with STxB alone or mixed with other adjuvants. To test whether this vaccine could break tolerance against self Ag, OVA transgenic mice were immunized with STxB-OVA alone or mixed with alpha-GalCer. Although no CTL induction was observed after immunization of OVA transgenic mice with STxB-OVA, tetramer assay clearly detected specific anti-OVA CD8(+) T cells in 8 of 11 mice immunized with STxB-OVA combined with alpha-GalCer. In addition, vaccination with STxB-OVA and alpha-GalCer conferred strong protection against a challenge with vaccinia virus encoding OVA with virus titers in the ovaries reduced by 5 log compared with nonimmunized mice. STxB combined with alpha-GalCer therefore appears as a promising vaccine strategy to more successfully establish protective CD8(+) T cell memory against intracellular pathogens and tumors.


Assuntos
Autoantígenos/imunologia , Galactosilceramidas/farmacologia , Tolerância Imunológica/efeitos dos fármacos , Toxinas Shiga/farmacologia , Vacinas Sintéticas/farmacologia , Vacínia/prevenção & controle , Animais , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Sinergismo Farmacológico , Galactosilceramidas/química , Galactosilceramidas/imunologia , Camundongos , Camundongos Transgênicos , Proteínas Oncogênicas Virais/química , Proteínas Oncogênicas Virais/imunologia , Ovalbumina/química , Ovalbumina/genética , Ovalbumina/imunologia , Proteínas E7 de Papillomavirus , Peptídeos/química , Peptídeos/imunologia , Peptídeos/farmacologia , Toxinas Shiga/química , Toxinas Shiga/imunologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Vacinas Sintéticas/química , Vacinas Sintéticas/imunologia , Vaccinia virus/imunologia
7.
Eur J Immunol ; 36(5): 1124-35, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16568496

RESUMO

The non-toxic B-subunit of Shiga toxin (STxB) interacts with the glycolipid Gb3, which is preferentially expressed on dendritic cells (DC) and B cells. After administration of STxB chemically coupled to OVA (STxB-OVA) in mice, we showed that the immunodominant OVA(257-264) peptide restricted by K(b) molecules is specifically presented by CD11c+ CD8alpha- DC, some of them displaying a mature phenotype. Using mice carrying a transgene encoding a diphtheria toxin receptor (DTR) under the control of the murine CD11c promoter, which allows inducible ablation of DC, we showed that DC are required for efficient priming of CTL after STxB-OVA vaccination. Immunization of mice with STxB-OVA induced OVA-specific CD8+ T cells detected ex vivo; these cells were long lasting, since they could be detected even 91 days after the last immunization and were composed of both central and memory T cells. Vaccination of mice with STxB-OVA and STxB coupled to E7, a protein derived from HPV16, inhibited tumor growth in prophylactic and therapeutic experiments. This effect was mainly mediated by CD8+ T cells. STxB therefore appears to be a powerful carrier directly targeting DC in vivo, resulting in a strong and durable CTL response associated with tumor protection.


Assuntos
Células Dendríticas/imunologia , Neoplasias Experimentais/imunologia , Ovalbumina/imunologia , Toxinas Shiga/farmacologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Feminino , Imunoterapia Adotiva , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/terapia , Fragmentos de Peptídeos/imunologia , Linfócitos T Citotóxicos/imunologia , Vacinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA