Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Negl Trop Dis ; 16(9): e0010779, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36170238

RESUMO

Amphotericin B is increasingly used in treatment of leishmaniasis. Here, fourteen independent lines of Leishmania mexicana and one L. infantum line were selected for resistance to either amphotericin B or the related polyene antimicrobial, nystatin. Sterol profiling revealed that, in each resistant line, the predominant wild-type sterol, ergosta-5,7,24-trienol, was replaced by other sterol intermediates. Broadly, two different profiles emerged among the resistant lines. Whole genome sequencing then showed that these distinct profiles were due either to mutations in the sterol methyl transferase (C24SMT) gene locus or the sterol C5 desaturase (C5DS) gene. In three lines an additional deletion of the miltefosine transporter gene was found. Differences in sensitivity to amphotericin B were apparent, depending on whether cells were grown in HOMEM, supplemented with foetal bovine serum, or a serum free defined medium (DM). Metabolomic analysis after exposure to AmB showed that a large increase in glucose flux via the pentose phosphate pathway preceded cell death in cells sustained in HOMEM but not DM, indicating the oxidative stress was more significantly induced under HOMEM conditions. Several of the lines were tested for their ability to infect macrophages and replicate as amastigote forms, alongside their ability to establish infections in mice. While several AmB resistant lines showed reduced virulence, at least two lines displayed heightened virulence in mice whilst retaining their resistance phenotype, emphasising the risks of resistance emerging to this critical drug.


Assuntos
Antiprotozoários , Leishmania mexicana , Camundongos , Animais , Anfotericina B/farmacologia , Leishmania mexicana/metabolismo , Nistatina , Soroalbumina Bovina/metabolismo , Esteróis , Estresse Oxidativo , Polienos , Transferases/metabolismo , Glucose , Ácidos Graxos Dessaturases/metabolismo , Antiprotozoários/farmacologia
2.
PLoS Negl Trop Dis ; 13(2): e0007052, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30716073

RESUMO

Amphotericin B is an increasingly important tool in efforts to reduce the global disease burden posed by Leishmania parasites. With few other chemotherapeutic options available for the treatment of leishmaniasis, the potential for emergent resistance to this drug is a considerable threat. Here we characterised four novel amphotericin B-resistant Leishmania mexicana lines. All lines exhibited altered sterol biosynthesis, and hypersensitivity to pentamidine. Whole genome sequencing demonstrated resistance-associated mutation of the sterol biosynthesis gene sterol C5-desaturase in one line. However, in three out of four lines, RNA-seq revealed loss of expression of sterol C24-methyltransferase (SMT) responsible for drug resistance and altered sterol biosynthesis. Additional loss of the miltefosine transporter was associated with one of those lines. SMT is encoded by two tandem gene copies, which we found to have very different expression levels. In all cases, reduced overall expression was associated with loss of the 3' untranslated region of the dominant gene copy, resulting from structural variations at this locus. Local regions of sequence homology, between the gene copies themselves, and also due to the presence of SIDER1 retrotransposon elements that promote multi-gene amplification, correlate to these structural variations. Moreover, in at least one case loss of SMT expression was not associated with loss of virulence in primary macrophages or in vivo. Whilst such repeat sequence-mediated instability is known in Leishmania genomes, its presence associated with resistance to a major antileishmanial drug, with no evidence of associated fitness costs, is a significant concern.


Assuntos
Anfotericina B/farmacologia , Instabilidade Genômica , Leishmania mexicana/efeitos dos fármacos , Leishmania mexicana/genética , Metiltransferases/genética , Animais , Antiprotozoários/farmacologia , Resistência a Medicamentos , Regulação Enzimológica da Expressão Gênica , Humanos , Metiltransferases/metabolismo
3.
PLoS One ; 7(3): e34416, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22470569

RESUMO

BACKGROUND: Guillain-Barré syndrome (GBS) is a post-infectious polyradiculoneuropathy, frequently associated with antecedent Campylobacter jejuni (C. jejuni) infection. The presence of sialic acid on C. jejuni lipo-oligosaccharide (LOS) is considered a risk factor for development of GBS as it crucially determines the structural homology between LOS and gangliosides, explaining the induction of cross-reactive neurotoxic antibodies. Sialylated C. jejuni are recognised by TLR4 and sialoadhesin; however, the functional implications of these interactions in vivo are unknown. METHODOLOGY/PRINCIPAL FINDINGS: In this study we investigated the effects of bacterial sialylation on phagocytosis and cytokine secretion by mouse myeloid cells in vitro and in vivo. Using fluorescently labelled GM1a/GD1a ganglioside-mimicking C. jejuni strains and corresponding (Cst-II-mutant) control strains lacking sialic acid, we show that sialylated C. jejuni was more efficiently phagocytosed in vitro by BM-MΦ, but not by BM-DC. In addition, LOS sialylation increased the production of IL-10, IL-6 and IFN-ß by both BM-MΦ and BM-DC. Subsequent in vivo experiments revealed that sialylation augmented the deposition of fluorescent bacteria in splenic DC, but not macrophages. In addition, sialylation significantly amplified the production of type I interferons, which was independent of pDC. CONCLUSIONS/SIGNIFICANCE: These results identify novel immune stimulatory effects of C. jejuni sialylation, which may be important in inducing cross-reactive humoral responses that cause GBS.


Assuntos
Infecções por Campylobacter/patologia , Campylobacter jejuni/metabolismo , Citocinas/metabolismo , Lipopolissacarídeos/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Fagocitose , Animais , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Gangliosídeos/metabolismo , Síndrome de Guillain-Barré/fisiopatologia , Interferon beta/metabolismo , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Baço/imunologia
4.
Eur J Immunol ; 38(5): 1238-46, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18398931

RESUMO

We demonstrate that uptake of oligomeric cognate antigen (OVA-hen egg lysozyme, OVA-HEL) alone or incorporated in immune-stimulating complexes (ISCOMS) facilitates presentation and simultaneous cross-presentation of OVA by HEL-specific B cells in vitro. HEL-specific B cells stimulated CD8(+) T cell responses in vitro to the same extent as bone marrow-derived dendritic cells. Cross-presentation by specific B cells required endosomal acidification, proteasomal processing and classical MHC class I/peptide transport. Specific B cells also acquired both antigens rapidly in vivo and presented them to CD4(+) T cells. However, only HEL-specific B cells from OVA-HEL ISCOMS-immunised mice could cross-present OVA to naive OVA-specific CD8(+) T cells. Antigen-specific B cells were also activated selectively by OVA-HEL ISCOMS in vitro and importantly, the presence of HEL-specific B cells promoted the persistence of clonal expansion of OVA-specific CD8(+) T cells after in vivo immunisation with OVA-HEL ISCOMS. These results demonstrate preferential MHC class I and class II processing of cognate antigen incorporated in ISCOMS by specific B cells in vitro and in vivo, highlighting the ability of ISCOMS to target B cells and offering novel insights into the role of B cells in cross-presentation to CD8(+) T cells.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos/imunologia , Linfócitos B/imunologia , Epitopos de Linfócito B/imunologia , ISCOMs , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Transferência Adotiva , Animais , Apresentação de Antígeno/efeitos dos fármacos , Antígenos/metabolismo , Linfócitos B/metabolismo , Brefeldina A/farmacologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/transplante , Proliferação de Células , Cloroquina/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Cinética , Linfonodos/citologia , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Mutantes , Camundongos Transgênicos , Muramidase/administração & dosagem , Muramidase/química , Muramidase/imunologia , Ovalbumina/administração & dosagem , Ovalbumina/química , Ovalbumina/imunologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Receptores de Antígenos de Linfócitos T/genética , Vacinação
5.
Vaccine ; 24(24): 5201-10, 2006 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-16650917

RESUMO

There is increasing concern over the efficacy of existing vaccines for diphtheria and there is interest in the development of a mucosally active formulation which might improve local protection. Lipophilic immune stimulating complexes (ISCOMS) containing Quil A are active by both parenteral and mucosal routes and here we have established methods for incorporating palmitified diphtheria toxoid (DT) into ISCOMS. The resulting formulation was immunogenic by the subcutaneous, oral and intranasal routes, with very low doses of DT inducing systemic humoral immune responses, as well as cell mediated immunity including both gammaIFN and IL5 production. Intranasal immunisation with DT in ISCOMS also stimulated significant local antibody production in tracheal washes, as well as cellular immunity in draining lymphoid tissues and serum neutralising antibodies. Finally, subcutaneous immunisation of guinea pigs with DT in ISCOMS primed protective immunity against challenge with diphtheria holotoxin more efficiently than the equivalent doses of DT in the conventional alum vaccine. ISCOMS based vaccines may provide a novel strategy for mucosal and systemic immunisation against diphtheria.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Toxoide Diftérico/administração & dosagem , ISCOMs/administração & dosagem , Administração Intranasal , Administração Oral , Animais , Anticorpos Antibacterianos/sangue , Feminino , Cobaias , Imunidade nas Mucosas , Imunização , Imunoglobulina G/sangue , Injeções Subcutâneas , Camundongos , Camundongos Endogâmicos BALB C
6.
J Immunol ; 176(6): 3697-706, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16517738

RESUMO

The cholera toxin A1 (CTA1)-DD/QuilA-containing, immune-stimulating complex (ISCOM) vector is a rationally designed mucosal adjuvant that greatly potentiates humoral and cellular immune responses. It was developed to incorporate the distinctive properties of either adjuvant alone in a combination that exerted additive enhancing effects on mucosal immune responses. In this study we demonstrate that CTA1-DD and an unrelated Ag can be incorporated together into the ISCOM, resulting in greatly augmented immunogenicity of the Ag. To demonstrate its relevance for protection against infectious diseases, we tested the vector incorporating PR8 Ag from the influenza virus. After intranasal immunization we found that the immunogenicity of the PR8 proteins were significantly augmented by a mechanism that was enzyme dependent, because the presence of the enzymatically inactive CTA1R7K-DD mutant largely failed to enhance the response over that seen with ISCOMs alone. The combined vector was a highly effective enhancer of a broad range of immune responses, including specific serum Abs and balanced Th1 and Th2 CD4(+) T cell priming as well as a strong mucosal IgA response. Unlike unmodified ISCOMs, Ag incorporated into the combined vector could be presented by B cells in vitro and in vivo as well as by dendritic cells; it also accumulated in B cell follicles of draining lymph nodes when given s.c. and stimulated much enhanced germinal center reactions. Strikingly, the enhanced adjuvant activity of the combined vector was absent in B cell-deficient mice, supporting the idea that B cells are important for the adjuvant effects of the combined CTA1-DD/ISCOM vector.


Assuntos
Adjuvantes Imunológicos , Antígenos/imunologia , Linfócitos B/imunologia , Toxina da Cólera/imunologia , ISCOMs/imunologia , Imunidade nas Mucosas/imunologia , Proteínas Recombinantes de Fusão/imunologia , Animais , Anticorpos/imunologia , Anticorpos/metabolismo , Toxina da Cólera/metabolismo , ISCOMs/metabolismo , Linfonodos/metabolismo , Camundongos , Proteínas Recombinantes de Fusão/metabolismo , Células Th1/imunologia , Células Th2/imunologia
7.
Novartis Found Symp ; 252: 291-302; discussion 302-5, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14609226

RESUMO

The intestinal immune system discriminates between invasive pathogens and antigens that are harmless, such as food proteins and commensal bacteria. The latter groups of antigens normally induce tolerance and a breakdown in this homeostatic process can lead to diseases such as coeliac disease or Crohn's disease. The nature ofthe intestinal immune response depends on how antigen is presented to CD4+ T cells by dendritic cells (DCs). Both oral tolerance and priming are influenced by the numbers and activation status of DCs in the gut and its draining lymphoid tissues, and our current work indicates that dietary proteins are taken up preferentially by DCs in the lamina propria of the small intestine. These then migrate to interact with antigen-specific CD4+ T cells in the mesenteric lymph node. In vivo and in vitro studies using purified lamina propria DCs suggest these may play a unique role in the regulation of intestinal immune responses. We propose that local DCs are the gatekeepers of the mucosal immune system, inducing tolerance under physiological conditions, but being sufficiently responsive to inflammatory stimuli to allow T cell priming and protective immunity when necessary. In addition, we will discuss evidence that adjuvant vectors such as ISCOMS may be effective mucosal vaccines due to an ability to activate intestinal DCs.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Tolerância Imunológica , Imunidade nas Mucosas/imunologia , Animais , Homeostase , Humanos , Imunofenotipagem , Camundongos
8.
Immunology ; 110(1): 95-104, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12941146

RESUMO

Immune stimulating complexes (ISCOMs) containing the saponin adjuvant Quil A are vaccine adjuvants that promote a wide range of immune responses in vivo, including delayed-type hypersensitivity (DTH) and the secretion of both T helper 1 (Th1) and Th2 cytokines. However, the antigen-presenting cell (APC) responsible for the induction of these responses has not been characterized. Here we have investigated the role of dendritic cells (DC), macrophages (Mphi) and B cells in the priming of antigen-specific CD4+ T cells in vitro by ISCOMs containing ovalbumin (OVA). OVA ISCOMs pulsed bone marrow (BM)-derived DC but not BM Mphi, nor naïve B cells prime resting antigen-specific CD4+ T cells, and this response is greatly enhanced if DC are activated with lipopolysaccharide (LPS). Of the APC found in the spleen, only DC had the capacity to prime resting antigen specific CD4+ T cells following exposure to OVA ISCOMs in vitro, while Mphi and B cells were ineffective. DC, but not B cells purified from the draining lymph nodes of mice immunized with OVA ISCOMs also primed resting antigen-specific CD4+ T cells in vitro, suggesting that DC are also critical in vivo. Using DC and T cells from interleukin (IL)-12 p40-/- mice, we also identified a crucial role for IL-12 in the priming of optimal CD4+ T cell responses by OVA ISCOMs. We suggest that DC are the principal APC responsible for the priming of CD4+ T cells by ISCOMs in vivo and that directed targeting of these vectors to DC may enhance their efficancy as vaccine adjuvants.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , ISCOMs/imunologia , Interleucina-12/imunologia , Adjuvantes Imunológicos , Animais , Apresentação de Antígeno/imunologia , Linfócitos B/imunologia , Células Cultivadas , Células Dendríticas/imunologia , Relação Dose-Resposta Imunológica , Feminino , Genes MHC da Classe II , Ativação Linfocitária/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/imunologia
9.
Immunology ; 109(3): 374-83, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12807483

RESUMO

Immune stimulating complexes (ISCOMS) containing the saponin adjuvant Quil A are vaccine adjuvants that induce a wide range of immune responses in vivo, including strong class I major histocompatibility complex (MHC)-restricted cytotoxic T-lymphocyte activity. However, the antigen-presenting cell responsible for the induction of these responses has not been characterized. Here we have investigated the role of dendritic cells (DC) in the priming of antigen-specific CD8+ T cells in vitro by ISCOMS containing ovalbumin. Resting bone marrow DC pulsed with ovalbumin ISCOMS efficiently prime resting CD8+ T cells through a mechanism that is transporter associated with antigen processing (TAP) dependent, but independent of CD40 ligation and CD4+ T-cell help. Lipopolysaccharide-induced maturation of DC markedly enhances their ability to prime CD8+ T cells through a mechanism which is also independent of CD4+ T-cell help, but is dependent on CD40 ligation. Furthermore, DC maturation revealed a TAP-independent mechanism of CD8+ T-cell priming. Our results also show that class I MHC-restricted presentation of ovalbumin in ISCOMS by DC is sensitive to chloroquine and brefeldin A but insensitive to lactacystin. We suggest that DC may be the principal antigen-presenting cells responsible for the priming of CD8+ T cells by ISCOMS in vivo and that targeting these vectors to activated DC may enhance their presentation via a novel pathway of class I antigen processing.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , ISCOMs/imunologia , Animais , Apresentação de Antígeno/efeitos dos fármacos , Apresentação de Antígeno/imunologia , Brefeldina A/farmacologia , Linfócitos T CD4-Positivos/imunologia , Antígenos CD40/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Cloroquina/farmacologia , Relação Dose-Resposta Imunológica , Feminino , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia
10.
Int Immunol ; 15(6): 711-20, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12750355

RESUMO

Immune-stimulating complexes (ISCOMS) are adjuvant vectors which are unusual in being able to prime both CD4(+) and CD8(+) T cells by parenteral and mucosal routes. However, their mode of action is unclear and to define better the cellular interactions involved we have studied the ability of ISCOMS containing ovalbumin (OVA) to prime TCR transgenic CD4(+) or CD8(+) T cells in vivo. Immunization with OVA ISCOMS caused activation and clonal expansion of CD4(+) and CD8(+) T cells in the T cell areas of the draining lymph nodes, followed by the migration of both CD4(+) and CD8(+) T cells into the B cell follicle. The T cells were primed to proliferate and secrete IFN-gamma after re-stimulation in vitro with the appropriate OVA peptide and CD8(+) T cell priming occurred in the absence of CD4(+) T cells. Increasing the number of dendritic cells (DC) in vivo with flt3 ligand augmented the expansion and activation of the OVA-specific T cells, particularly CD8(+) T cells. These studies indicate DC play a central role in the priming of both CD4(+) and CD8(+) T cells in vivo, and suggest that an ability to target DC may allow ISCOMS to be powerful vaccine vectors for stimulating protective immunity.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , ISCOMs/farmacologia , Ativação Linfocitária/imunologia , Animais , Complexo Antígeno-Anticorpo/imunologia , Complexo Antígeno-Anticorpo/farmacologia , Feminino , ISCOMs/imunologia , Imunização , Camundongos , Ovalbumina/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...