Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Immunol Cell Biol ; 102(4): 235-239, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37921552

RESUMO

In the unique landscape of immunology research in New Zealand, this article explores the collaborative networks spanning the two main islands, through a conversation with Associate Professor Joanna Kirman and Dr Robert Weinkove. The discussions delve into their dynamic collaborations with countries such as Asia, Australia and the United States, from their laboratories at the University of Otago and the Malaghan Institute of Medical Research, respectively, provides insight into the translational research landscape of New Zealand, and the integration of Maori culture into all aspects of scientific research and clinical practise. Kirman's work in understanding immunological memory in tuberculosis and Weinkove's research in cancer immunotherapies, particularly CAR-T cells, are highlighted. The natural beauty and accessibility of New Zealand supports its research diversity.


Assuntos
Povos Aborígenes Australianos e Ilhéus do Estreito de Torres , Povo Maori , Humanos , Nova Zelândia , Austrália
2.
Nanomaterials (Basel) ; 11(8)2021 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-34443891

RESUMO

Currently available vaccines fail to provide consistent protection against tuberculosis (TB). New, improved vaccines are urgently needed for controlling the disease. The mycobacterial antigen fusions H4 (Ag85B-TB10.4) and H28 (Ag85B-TB10.4-Rv2660c) have been shown to be very immunogenic and have been considered as potential candidates for TB vaccine development. However, soluble protein vaccines are often poorly immunogenic, but augmented immune responses can be induced when selected antigens are delivered in particulate form. This study investigated whether the mycobacterial antigen fusions H4 and H28 can induce protective immunity when assembled into particulate vaccines (polyester nanoparticle-H4, polyester nanoparticle-H28, H4 nanoparticles and H28 nanoparticles). The particulate mycobacterial vaccines were assembled inside an engineered endotoxin-free production strain of Escherichia coli at high yield. Vaccine nanoparticles were purified and induced long-lasting antigen-specific T cell responses and protective immunity in mice challenged by aerosol with virulent Mycobacterium tuberculosis. A significant reduction of M. tuberculosis CFU, up to 0.7-log10 protection, occurred in the lungs of mice immunized with particulate vaccines in comparison to placebo-vaccinated mice (p < 0.0001). Polyester nanoparticles displaying the mycobacterial antigen fusion H4 induced a similar level of protective immunity in the lung when compared to M. bovis bacillus Calmette-Guérin (BCG), the currently approved TB vaccine. The safe and immunogenic polyester nanoparticle-H4 vaccine is a promising subunit vaccine candidate, as it can be cost-effectively manufactured and efficiently induces protection against TB.

3.
Immunol Cell Biol ; 99(7): 767-781, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33866609

RESUMO

Mycobacterium tuberculosis (Mtb) remains a global epidemic despite the widespread use of Bacillus Calmette-Guérin (BCG). Consequently, novel vaccines are required to facilitate a reduction in Mtb morbidity and mortality. PilVax is a peptide delivery strategy for the generation of highly specific mucosal immune responses and is based on the food-grade bacterium Lactococcus lactis that is used to express selected peptides engineered within the Streptococcus pyogenes M1T1 pilus, allowing for peptide amplification, stabilization and enhanced immunogenicity. In the present study, the dominant T-cell epitope from the Mtb protein Ag85B was genetically engineered into the pilus backbone subunit and expressed on the surface of L. lactis. Western blot and flow cytometry confirmed formation of pilus containing the peptide DNA sequence. B-cell responses in intranasally vaccinated mice were analyzed by ELISA while T-cell responses were analyzed by flow cytometry. Serum titers of peptide-specific immunoglobulin (Ig) G and IgA were detected, confirming that vaccination produced antibodies against the cognate peptide. Peptide-specific IgA was also detected across several mucosal sites sampled. Peptide-specific CD4+ T cells were detected at levels similar to those of mice immunized with BCG. PilVax immunization resulted in an unexpected increase in the numbers of CD3+ CD4- CD8- [double negative (DN)] T cells in the lungs of vaccinated mice. Analysis of cytokine production following stimulation with the cognate peptide showed the major cytokine producing cells to be CD4+ T cells and DN T cells. This study provides insight into the antibody and peptide-specific cellular immune responses generated by PilVax vaccination and demonstrates the suitability of this vaccine for conducting a protection study.


Assuntos
Lactococcus lactis , Mycobacterium tuberculosis , Vacinas contra a Tuberculose , Animais , Antígenos de Bactérias , Vacina BCG , Imunização , Camundongos , Peptídeos , Linfócitos T , Vacinação
5.
Cell Rep ; 28(12): 3061-3076.e5, 2019 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-31533031

RESUMO

Mucosal-associated invariant T (MAIT) cells can be activated via either their T cell receptor (TCR), which recognizes MR1-bound pyrimidines derived from microbial riboflavin biosynthesis, or via cytokines. These two modes of activation may act in concert or independently, depending upon the stimulus. It is unknown, however, how MAIT cell responses differ with the mode of activation. Here, we define transcriptional and effector responses of human CD8+ MAIT cells to TCR and cytokine stimulation. We report that MAIT cells rapidly respond to TCR stimulation, producing multiple cytokines and chemokines, altering their cytotoxic granule content and transcription factor expression, and upregulating co-stimulatory proteins. In contrast, cytokine-mediated activation is slower and results in a more limited response. Therefore, we propose that, in infections by riboflavin-synthesizing bacteria, MAIT cells play a key early role in effecting and coordinating immune responses, while in the absence of TCR stimulation, their role is likely to differ.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citocinas/imunologia , Ativação Linfocitária , Células T Invariantes Associadas à Mucosa/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Adulto , Linfócitos T CD8-Positivos/citologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Células T Invariantes Associadas à Mucosa/citologia
6.
Immunol Cell Biol ; 97(7): 615-616, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31283852
7.
Immunol Cell Biol ; 97(7): 647-655, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31141205

RESUMO

Tuberculosis (TB) is a serious infectious disease caused by infection with Mycobacterium tuberculosis, and kills more people annually than any other single infectious agent. Although a vaccine is available, it is only moderately effective and an improved vaccine is urgently needed. The ability to develop a more effective vaccine has been thwarted by a lack of understanding of the mechanism of vaccine-induced immune protection. Over recent decades, many novel TB vaccines have been developed and almost all have aimed to generate memory CD4 T cells. In this review, we critically evaluate evidence in the literature that supports the contention that memory CD4 T cells are the prime mediators of vaccine-induced protection against TB. Because of the lack of robust evidence supporting memory CD4 T cells in this role, the potential for B-cell antibody and "trained" innate cells as alternative mediators of protective immunity is explored.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Memória Imunológica , Mycobacterium tuberculosis/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Tuberculose/imunologia , Tuberculose/microbiologia , Imunidade Adaptativa , Animais , Anticorpos Antibacterianos/imunologia , Especificidade de Anticorpos/imunologia , Antígenos de Bactérias/imunologia , Vacina BCG/administração & dosagem , Vacina BCG/efeitos adversos , Vacina BCG/imunologia , Glicosilação , Humanos , Imunidade Inata , Isotipos de Imunoglobulinas/imunologia , Isotipos de Imunoglobulinas/metabolismo , Avaliação de Resultados em Cuidados de Saúde , Subpopulações de Linfócitos T/citologia , Tuberculose/prevenção & controle , Vacinas contra a Tuberculose/administração & dosagem , Vacinas contra a Tuberculose/efeitos adversos , Vacinas contra a Tuberculose/imunologia , Vacinação/efeitos adversos , Vacinação/métodos
8.
Front Microbiol ; 10: 402, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30906286

RESUMO

Lung infection by Mycobacterium tuberculosis is characterized by chronic infection of lung-resident macrophages, long considered to be the primary hosts and determinants of the outcome of the early immune response. Although alveolar macrophages are well-known to host intracellular mycobacteria at later stages of disease, little is known about the earliest events of the innate immune response. The phagocytes that take up mycobacteria immediately following infection, and how the early lung phagocyte response is altered by vaccination with M. bovis bacille Calmette-Guérin (BCG) were unknown. Using BCG expressing the bright red fluorescent protein tdTomato and flow cytometry, we modeled early infection in C57BL/6 mice and tracked phagocyte population kinetics and uptake of mycobacteria, to better understand the involvement of specific phagocyte subsets. By 1 day post-infection, dose-dependent accumulation of neutrophils was observed and surprisingly, granulocytes comprised a greater proportion of infected phagocytes than alveolar macrophages. By 7 days post-infection alveolar macrophages had become the dominant BCG-associated phagocytes. Prior mucosal BCG exposure provided immunized mice with greater frequencies and numbers of lung macrophage subsets, and a significantly greater proportion of alveolar macrophages expressed CD11b prior to and following challenge infection. These data provide the first evidence of granulocytes as the dominant infected phagocyte subset early after mycobacterial infection, and highlight enhanced recruitment of lung macrophages as a factor associated with protection in BCG-immunized mice.

9.
FASEB J ; 33(6): 7505-7518, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30870010

RESUMO

A major obstacle to tuberculosis (TB)-subunit-vaccine development has been the induction of inadequate levels of protective immunity due to the limited breadth of antigen in vaccine preparations. In this study, immunogenic mycobacterial fusion peptides Ag85B-TB10.4 and Ag85B-TB10.4-Rv2660c were covalently displayed on the surface of self-assembled polyester particles. This study investigated whether polyester particles displaying mycobacterial antigens could provide augmented immunogenicity (i.e., offer an innovative vaccine formulation) when compared with free soluble antigens. Herein, polyester particle-based particulate vaccines were produced in an endotoxin-free Escherichia coli strain and emulsified with the adjuvant dimethyl dioctadecyl ammonium bromide. C57BL/6 mice were used to study the immunogenicity of formulated particulate vaccines. The result of humoral immunity showed the antibodies only interacted with target antigens and not with PhaC and the background proteins of the production host. The analysis of T helper 1 cellular immunity indicated that a relatively strong production of cellular immunity biomarkers, IFN-γ and IL-17A cytokines, was induced by particulate vaccines when compared with the respective soluble controls. This study demonstrated that polyester particles have the potential to perform as a mycobacterial antigen-delivery agent to induce augmented antigen-specific immune responses in contrast to free soluble vaccines.-Chen, S., Sandford, S., Kirman, J. R., Rehm, B. H. A. Innovative antigen carrier system for the development of tuberculosis vaccines.


Assuntos
Antígenos/administração & dosagem , Portadores de Fármacos , Vacinas contra a Tuberculose/administração & dosagem , Animais , Meios de Cultura , Citocinas/metabolismo , Escherichia coli/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium tuberculosis/imunologia
10.
Front Immunol ; 9: 953, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29867941

RESUMO

Bloodstream infections induce considerable morbidity, high mortality, and represent a significant burden of cost in health care; however, our understanding of the immune response to bacteremia is incomplete. Langerin+ CD8α+ dendritic cells (DCs), residing in the marginal zone of the murine spleen, have the capacity to cross-prime CD8+ T cells and produce IL-12, both of which are important components of antimicrobial immunity. Accordingly, we hypothesized that this DC subset may be a key promoter of adaptive immune responses to blood-borne bacterial infections. Utilizing mice that express the diphtheria toxin receptor under control of the langerin promoter, we investigated the impact of depleting langerin+ CD8α+ DCs in a murine model of intravenous infection with Mycobacterium bovis bacille Calmette-Guerin (BCG). In the absence of langerin+ CD8α+ DCs, the immune response to blood-borne BCG infection was diminished: bacterial numbers in the spleen increased, serum IL-12p40 decreased, and delayed CD8+ T cell activation, proliferation, and IFN-γ production was evident. Our data revealed that langerin+ CD8α+ DCs play a pivotal role in initiating CD8+ T cell responses and IL-12 production in response to bacteremia and may influence the early control of systemic bacterial infections.


Assuntos
Antígenos de Superfície/metabolismo , Infecções Bacterianas/etiologia , Infecções Bacterianas/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Ativação Linfocitária/imunologia , Lectinas de Ligação a Manose/metabolismo , Animais , Carga Bacteriana , Modelos Animais de Doenças , Interleucina-12/biossíntese , Subunidade p40 da Interleucina-12/sangue , Depleção Linfocítica , Masculino , Camundongos , Mycobacterium bovis/imunologia , Baço/imunologia , Baço/metabolismo , Baço/microbiologia , Baço/patologia , Especificidade do Receptor de Antígeno de Linfócitos T
11.
Immunol Cell Biol ; 96(4): 379-389, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29363172

RESUMO

The tuberculosis (TB) vaccine bacille Calmette-Guérin (BCG) prevents disseminated childhood TB; however, it fails to protect against the more prevalent pulmonary TB. Limited understanding of the immune response to Mycobacterium tuberculosis, the causative agent of TB, has hindered development of improved vaccines. Although memory CD4 T cells are considered the main mediators of protection against TB, recent studies suggest there are other key subsets that contribute to antimycobacterial immunity. To that end, innate cells may be involved in the protective response. In this study, we investigated the primary response of innate lymphoid cells (ILCs) to BCG exposure. Using a murine model, we showed that ILCs increased in number in the lungs and lymph nodes in response to BCG vaccination. Additionally, there was significant production of the antimycobacterial cytokine IFN-γ by ILCs. As ILCs are located at mucosal sites, it was investigated whether mucosal vaccination (intranasal) stimulated an enhanced response compared to the traditional vaccination approach (intradermal or subcutaneous). Indeed, in response to intranasal vaccination, the number of ILCs, and IFN-γ production in NK cells and ILC1s in the lungs and lymph nodes, were higher than that provoked through intradermal or subcutaneous vaccination. This work provides the first evidence that BCG vaccination activates ILCs, paving the way for future research to elucidate the protective potential of ILCs against mycobacterial infection. Additionally, the finding that lung ILCs respond rigorously to mucosal vaccination may have implications for the delivery of novel TB vaccines.


Assuntos
Vacina BCG/imunologia , Imunidade Inata , Pulmão/citologia , Linfócitos/citologia , Linfócitos/imunologia , Vacinação , Animais , Biomarcadores/metabolismo , Contagem de Células , Linfonodos/citologia , Linfonodos/imunologia , Masculino , Camundongos Endogâmicos C57BL , Mucosa/imunologia , Fenótipo
12.
Microbiol Spectr ; 4(6)2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-28087940

RESUMO

Immunological memory is a central feature of the adaptive immune system and a prerequisite for generating effective vaccines. Understanding long-term memory responses to Mycobacterium tuberculosis will thus provide us with valuable insights that can guide us in the search for a novel vaccine against tuberculosis (TB). For many years, triggering CD4 T cells and, in particular, those secreting interferon-γ has been the goal of most TB vaccine research, and numerous data from animals and humans support the key role of this subset in protective immunity. More recently, we have learned that the memory response required for effective control of M. tuberculosis is much more complex, probably involving several phenotypically different CD4 T cell subsets as well as other cell types that are yet to be defined. Herein, we describe recent insights into memory immunity to TB in the context of both animal models and the human infection. With the increasing amount of data generated from clinical testing of novel TB vaccines, we also summarize recent knowledge of vaccine-induced memory immunity.


Assuntos
Imunidade Adaptativa , Memória Imunológica , Tuberculose/imunologia , Tuberculose/prevenção & controle , Animais , Modelos Animais de Doenças , Humanos , Vacinas contra a Tuberculose/imunologia
13.
Oncoimmunology ; 4(10): e1042199, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26451307

RESUMO

Local immune-activating therapies seek to improve the presentation of tumor antigen, thereby promoting the activation of antitumor CD8+ T cells and delaying tumor growth. Surprisingly, little is known about the ability of these therapies to stimulate antitumor CD4+ T cells. We examined tumor-specific CD4+ T cell responses after peri-tumoral administration of the TLR3 agonist polyinosinic-polycytidylic acid (poly I:C), or the danger signal monosodium urate crystals in combination with Mycobacterium smegmatis (MSU + Msmeg) in mice. Both treatments delayed tumor growth, however, only MSU + Msmeg induced proliferation of tumor-specific CD4+ T cells in the draining lymph node (dLN). In line with the proliferation data, administration of MSU + Msmeg, but not poly I:C, enhanced the infiltration of CD4+FoxP3- T cells into the tumor, increased their capacity to produce IFNγ and TNF-α, and decreased PD-1 expression on tumor-infiltrating CD8+ T cells. Induction of CD4+ T cell proliferation by treatment with MSU + Msmeg required IL-1ßR signaling, as it was blocked by administration of the IL-1ßR antagonist Anakinra. In addition, treatment with Anakinra or with anti-CD4 also reversed the increased survival after tumor challenge in MSU + Msmeg treated mice. Thus, peri-tumoral treatment with MSU + Msmeg results in IL-1ßR-dependent priming of antitumor CD4+ T cells in the LN, with consequent superior activation of CD4+ and CD8+ T cells within the tumor, and sustained antitumor activity.

14.
J Immunol Methods ; 406: 104-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24556588

RESUMO

Splenic langerin(+) CD8α(+) dendritic cells (DCs) have exhibited a critical role in cross-priming CD8(+) T cell responses. To further study the roles of this DC subset, a protocol for the continuous depletion of langerin(+) CD8α(+) DCs was established using the pre-existing lang-DTREGFP mouse model. Due to the fast turnover rate of splenic CD8α(+) DCs, maintaining the depletion of langerin(+) CD8α(+) DCs required multiple diphtheria toxin (DT) treatments. We found that prolonged treatment with DT did not cause weight loss, or neutrophilia, as reported in some DT-based depletion models. Therefore, the in vivo depletion of murine langerin(+) CD8α(+) DCs can be maintained over time to analyse their function during the full course of an immune response.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Toxina Diftérica/imunologia , Depleção Linfocítica , Animais , Apresentação de Antígeno/imunologia , Antígenos de Superfície/imunologia , Antígenos CD8/imunologia , Toxina Diftérica/administração & dosagem , Lectinas Tipo C/imunologia , Ativação Linfocitária/imunologia , Lectinas de Ligação a Manose/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Baço/citologia , Baço/imunologia
15.
J Immunol ; 191(4): 1984-92, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23858033

RESUMO

Local treatment with selected TLR ligands or bacteria such as bacillus Calmette-Guérin increases antitumor immune responses and delays tumor growth. It is thought that these treatments may act by activating tumor-associated dendritic cells (DCs), thereby supporting the induction of antitumor immune responses. However, common parameters of successful immune activation have not been identified. We used mouse models to compare treatments with different immune-activating agents for the ability to delay tumor growth, improve priming of tumor-specific T cells, and induce early cytokine production and DC activation. Treatment with polyinosinic-polycytidylic acid or a combination of monosodium urate crystals and Mycobacterium smegmatis was effective at delaying the growth of s.c. B16 melanomas, orthotopic 4T1 mammary carcinomas, and reducing 4T1 lung metastases. In contrast, LPS, monosodium urate crystals, or M. smegmatis alone had no activity. Effective treatments required both NK1.1(+) and CD8(+) cells, and resulted in increased T cell priming and the infiltration of NK cells and CD8(+) T cells in tumors. Unexpectedly, both effective and ineffective treatments increased DC numbers and the expression of costimulatory molecules in the tumor-draining lymph node. However, only effective treatments induced the rapid appearance of a population of monocyte-derived DCs in the draining lymph node, early release of IL-12p70 and IFN-γ, and low IL-10 in the serum. These results suggest that the activation of existing DC subsets is not sufficient for the induction of antitumor immune responses, whereas early induction of Th1 cytokines and monocyte-derived DCs are features of successful activation of antitumor immunity.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Carcinoma/terapia , Células Dendríticas/imunologia , Imunoterapia Adotiva , Imunoterapia , Células Matadoras Naturais/imunologia , Linfócitos do Interstício Tumoral/imunologia , Neoplasias Mamárias Experimentais/terapia , Melanoma Experimental/terapia , Monócitos/imunologia , Mycobacterium smegmatis/imunologia , Poli I-C/farmacologia , Subpopulações de Linfócitos T/imunologia , Ácido Úrico/farmacologia , Imunidade Adaptativa , Animais , Terapia Biológica , Carcinoma/imunologia , Carcinoma/secundário , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Citocinas/biossíntese , Citocinas/genética , Escherichia coli/imunologia , Feminino , Interferon gama/metabolismo , Interleucina-10/sangue , Interleucina-12/metabolismo , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Linfonodos/patologia , Masculino , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Melanoma Experimental/imunologia , Melanoma Experimental/secundário , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Monócitos/efeitos dos fármacos , Ovalbumina/imunologia , Fragmentos de Peptídeos/imunologia , Poli I-C/uso terapêutico , Quimera por Radiação , Receptores de Antígenos de Linfócitos T/imunologia , Subpopulações de Linfócitos T/transplante , Células Th1/imunologia , Células Th1/metabolismo , Ácido Úrico/uso terapêutico
16.
J Microbiol ; 51(5): 651-8, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23800952

RESUMO

Many whole cell screens of chemical libraries currently in use are based on inhibition of bacterial growth. The goal of this study was to develop a chemical library screening model that enabled detection of compounds that are active against drug-tolerant non-growing cultures of Mycobacterium tuberculosis. An in vitro model of low metabolically active mycobacteria was established with 8 and 30 day old cultures of M. smegmatis and M. tuberculosis, respectively. Reduction of resazurin was used as a measure of viability and the assay was applied in screens of chemical libraries for bactericidal compounds. The model provided cells that were phenotypically-resilient to killing by first and second-line clinical drugs including rifampicin. Screening against chemical libraries identified proteasome inhibitors, NSC310551 and NSC321206, and a structurally-related series of thiosemicarbazones, as having potent killing activity towards aged cultures. The inhibitors were confirmed as active against virulent M. tuberculosis strains including multi- and extensively-drug resistant clinical isolates. Our library screen enabled detection of compounds with a potent level of bactericidal activity towards phenotypically drug-tolerant cultures of M. tuberculosis.


Assuntos
Antituberculosos/isolamento & purificação , Antituberculosos/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Tolerância a Medicamentos , Viabilidade Microbiana/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/fisiologia , Humanos , Mycobacterium smegmatis/efeitos dos fármacos , Mycobacterium smegmatis/fisiologia , Oxazinas/metabolismo , Oxirredução , Coloração e Rotulagem/métodos , Xantenos/metabolismo
17.
Tuberculosis (Edinb) ; 93(2): 115-22, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23167967

RESUMO

Anti-mycobacterial immunity is guided by specialised antigen presenting cells known as dendritic cells, which are essential for both initiating and maintaining T cell immune responses during infection. The dendritic cell population can be divided into functionally distinct subsets that differ in their ability to present antigen and produce key TH1 cytokines, such as IL-12. This review discusses recent studies, in murine models, investigating which dendritic cell populations are important for mycobacterial control.


Assuntos
Células Dendríticas/imunologia , Infecções por Mycobacterium/imunologia , Mycobacterium/crescimento & desenvolvimento , Linfócitos T/imunologia , Animais , Apresentação de Antígeno/imunologia , Modelos Animais de Doenças , Interleucina-12/biossíntese , Camundongos , Infecções por Mycobacterium/microbiologia
18.
PLoS One ; 7(9): e45888, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23049885

RESUMO

Oral delivery of BCG in a lipid formulation (Liporale™-BCG) targets delivery of viable bacilli to the mesenteric lymph nodes and confers protection against an aerosol Mycobacterium tuberculosis challenge. The magnitude, quality and duration of the effector and memory immune response induced by Liporale™-BCG vaccination is unknown. Therefore, we compared the effector and memory CD4(+) T cell response in the spleen and lungs of mice vaccinated with Liporale™-BCG to the response induced by subcutaneous BCG vaccination. Liporale™-BCG vaccination induced a long-lived CD4(+) T cell response, evident by the detection of effector CD4(+) T cells in the lungs and a significant increase in the number of Ag85B tetramer-specific CD4(+) T cells in the spleen up to 30 weeks post vaccination. Moreover, following polyclonal stimulation, Liporale™-BCG vaccination, but not s.c. BCG vaccination, induced a significant increase in both the percentage of CD4(+) T cells in the lungs capable of producing IFNγ and the number of multifunctional CD4(+) T cells in the lungs at 30 weeks post vaccination. These results demonstrate that orally delivered Liporale™-BCG vaccine induces a long-lived multifunctional immune response, and could therefore represent a practical and effective means of delivering novel BCG-based TB vaccines.


Assuntos
Vacina BCG/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Vacinas/uso terapêutico , Aciltransferases/química , Administração Oral , Animais , Antígenos de Bactérias/química , Proteínas de Bactérias/química , Feminino , Sistema Imunitário , Interferon gama/metabolismo , Lipídeos/química , Pulmão/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium tuberculosis/metabolismo , Baço/citologia
19.
Tuberculosis (Edinb) ; 92(5): 422-33, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22738879

RESUMO

Several studies have used adoptive transfer of purified T cell subsets into immunodeficient mice to determine the subset of T cells responsible for mediating protection against Mycobacterium tuberculosis. These studies suggested that CD62L(hi) memory CD4(+) T cells from BCG-vaccinated mice are key for protection against tuberculosis. Importantly, we observed that transfer of naïve CD4(+) T cells into Rag1-/- recipients protected against a mycobacterial challenge as well as transfer of BCG-experienced CD4(+) T cells. We found that transfer of total CD4(+) T cells from naïve mice or enriched CD62L(hi)CD4(+) T cells from BCG-vaccinated mice into Rag1-/- recipients induced severe colitis by 3 weeks post cell transfer, whereas transfer of CD62L(lo)CD4(+) T cells from BCG-vaccinated mice did not. Naïve and CD62L(hi)CD4(+) T cells proliferated extensively upon transfer and developed an activated effector phenotype in the lung, even in the absence of infectious challenge. The induction of colitis and systemic cytokine response induced by the transfer and subsequent activation of CD4(+) T cells from naïve mice or CD62L(hi)CD4(+) T cells from BCG-vaccinated mice, into immunodeficient recipients, may heighten their ability to protect against mycobacterial challenge. This raises doubts about the validity of this model to study CD4(+) T cell-mediated protection against tuberculosis.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Colite/imunologia , Colo/imunologia , Mycobacterium bovis/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Transferência Adotiva , Animais , Antígenos de Bactérias/imunologia , Colite/patologia , Colo/patologia , Citometria de Fluxo , Proteínas de Homeodomínio/imunologia , Memória Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Modelos Imunológicos , Mycobacterium bovis/patogenicidade , Mycobacterium tuberculosis/patogenicidade , Tuberculose/patologia , Vacinação
20.
Cancer Immunol Immunother ; 61(12): 2333-42, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22714285

RESUMO

Mycobacteria and their cell wall components have been used with varying degrees of success to treat tumors, and Mycobacterium bovis BCG remains in use as a standard treatment for superficial bladder cancer. Mycobacterial immunotherapy is very effective in eliciting local immune responses against solid tumors when administered topically; however, its effectiveness in eliciting adaptive immune responses has been variable. Using a subcutaneous mouse thymoma model, we investigated whether immunotherapy with Mycobacterium smegmatis, a fast-growing mycobacterium of low pathogenicity, induces a systemic adaptive immune response. We found that M. smegmatis delivered adjacent to the tumor site elicited a systemic anti-tumor immune response that was primarily mediated by CD8(+) T cells. Of note, we identified a CD11c(+)CD40(int)CD11b(hi)Gr-1(+) inflammatory DC population in the tumor-draining lymph nodes that was found only in mice treated with M. smegmatis. Our data suggest that, rather than rescuing the function of the DC already present in the tumor and/or tumor-draining lymph node, M. smegmatis treatment may promote anti-tumor immune responses by inducing the involvement of a new population of inflammatory cells with intact function.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Mycobacterium smegmatis/imunologia , Timoma/imunologia , Timoma/terapia , Neoplasias do Timo/imunologia , Neoplasias do Timo/terapia , Imunidade Adaptativa/imunologia , Animais , Imunoterapia/métodos , Inflamação/imunologia , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...