Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Sci Rep ; 9(1): 17401, 2019 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-31758014

RESUMEN

Vaccination is the most effective method to prevent infectious diseases. However, approaches to identify novel vaccine candidates are commonly laborious and protracted. While surface proteins are suitable vaccine candidates and can elicit antibacterial antibody responses, systematic approaches to define surfomes from gram-negatives have rarely been successful. Here we developed a combined discovery-driven mass spectrometry and computational strategy to identify bacterial vaccine candidates and validate their immunogenicity using a highly prevalent gram-negative pathogen, Helicobacter pylori, as a model organism. We efficiently isolated surface antigens by enzymatic cleavage, with a design of experiment based strategy to experimentally dissect cell surface-exposed from cytosolic proteins. From a total of 1,153 quantified bacterial proteins, we thereby identified 72 surface exposed antigens and further prioritized candidates by computational homology inference within and across species. We next tested candidate-specific immune responses. All candidates were recognized in sera from infected patients, and readily induced antibody responses after vaccination of mice. The candidate jhp_0775 induced specific B and T cell responses and significantly reduced colonization levels in mouse therapeutic vaccination studies. In infected humans, we further show that jhp_0775 is immunogenic and activates IFNγ secretion from peripheral CD4+ and CD8+ T cells. Our strategy provides a generic preclinical screening, selection and validation process for novel vaccine candidates against gram-negative bacteria, which could be employed to other gram-negative pathogens.


Asunto(s)
Vacunas Bacterianas , Bacterias Gramnegativas , Espectrometría de Masas , Antígenos Bacterianos/química , Antígenos Bacterianos/inmunología , Antígenos de Superficie/química , Antígenos de Superficie/inmunología , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Vacunas Bacterianas/química , Vacunas Bacterianas/inmunología , Biología Computacional/métodos , Bacterias Gramnegativas/inmunología , Proteómica/métodos , Reproducibilidad de los Resultados
2.
Sci Rep ; 7(1): 13636, 2017 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-29057967

RESUMEN

Helicobacter pylori γ-glutamyl transferase (gGT) is a key bacterial virulence factor that is not only important for bacterial gastric colonization but also related to the development of gastric pathology. Despite accumulating evidence for pathogenic and immunologic functions of H. pylori gGT, it is still unclear how it supports gastric colonization and how its specific effects on the host's innate and adaptive immune responses contribute to colonization and pathology. We have compared mice showing similar bacterial load after infection with gGT-proficient or gGT-deficient H. pylori to analyse the specific role of the enzyme during infection. Our data indicate that H. pylori gGT supports initial colonization. Nevertheless, bacteria lacking gGT can still colonize and persist. We observed that the presence of gGT during infection favoured a proinflammatory innate and adaptive immune response. Notably, H. pylori gGT activity was linked to increased levels of IFNγ, which were attributed to a differential recruitment of CD8+ T cells to the stomach. Our data support an essential role for H. pylori gGT in gastric colonization and further suggest that gGT favours infiltration of CD8+ cells to the gastric mucosa, which might play an important and yet overlooked role in the pathogenesis of H. pylori.


Asunto(s)
Proteínas Bacterianas/metabolismo , Linfocitos T CD8-positivos/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter pylori/enzimología , Factores de Virulencia/metabolismo , gamma-Glutamiltransferasa/metabolismo , Inmunidad Adaptativa , Animales , Linfocitos T CD8-positivos/enzimología , Linfocitos T CD8-positivos/microbiología , Modelos Animales de Enfermedad , Femenino , Infecciones por Helicobacter/enzimología , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/patología , Helicobacter pylori/crecimiento & desarrollo , Inmunidad Innata , Interferón gamma/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Estómago/enzimología , Estómago/inmunología , Estómago/microbiología , Estómago/patología
3.
Gut ; 66(8): 1369-1381, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-27196595

RESUMEN

OBJECTIVE: Lymphotoxin ß receptor (LTßR) signalling has been implicated in inflammation-associated tumour development in different tissues. We have analysed the role of LTßR and alternative NF-κB signalling in Helicobacter pylori-mediated gastric inflammation and pathology. DESIGN: We analysed several ligands and receptors of the alternative NF-κB pathway, RelB, p52 nuclear translocation and target genes in tissue samples of H. pylori-infected patients with different degrees of gastritis or early gastric tumours by in situ hybridisation, immunohistochemistry, Western blot and real-time PCR analyses. Molecular mechanisms involved in LTßR activation by H. pylori were assessed in vitro using human gastric cancer cell lines and distinct H. pylori isolates. The effects of blocking or agonistically activating LTßR on gastric pathology during challenge with a human pathogenic H. pylori strain were studied in a mouse model. RESULTS: Among the tested candidates, LT was significantly increased and activated alternative NF-κB signalling was observed in the gastric mucosa of H. pylori-infected patients. H. pyloriinduced LTßR-ligand expression in a type IV secretion system-dependent but CagA-independent manner, resulting in activation of the alternative NF-κB pathway, which was further enhanced by blocking canonical NF-κB during infection. Blocking LTßR signalling in vivo suppressed H. pylori-driven gastritis, whereas LTßR activation in gastric epithelial cells of infected mice induced a broadened pro-inflammatory chemokine milieu, resulting in exacerbated pathology. CONCLUSIONS: LTßR-triggered activation of alternative NF-κB signalling in gastric epithelial cells executes H. pylori-induced chronic gastritis, representing a novel target to restrict gastric inflammation and pathology elicited by H. pylori, while exclusively targeting canonical NF-κB may aggravate pathology by enhancing the alternative pathway.


Asunto(s)
Quimiocinas/metabolismo , Gastritis/metabolismo , Infecciones por Helicobacter/metabolismo , Helicobacter pylori , Receptor beta de Linfotoxina/metabolismo , FN-kappa B/metabolismo , Sistemas de Secreción Tipo IV/metabolismo , Animales , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Línea Celular Tumoral , Quimiocina CCL2/metabolismo , Quimiocina CCL20/metabolismo , Quimiocina CXCL10/metabolismo , Células Epiteliales/metabolismo , Femenino , Mucosa Gástrica/metabolismo , Gastritis/microbiología , Infecciones por Helicobacter/complicaciones , Humanos , Receptor beta de Linfotoxina/antagonistas & inhibidores , Receptor beta de Linfotoxina/genética , Ratones , Ratones Endogámicos C57BL , ARN Mensajero , Transducción de Señal , Factor de Transcripción ReIB/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
4.
PLoS One ; 11(5): e0154643, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27138472

RESUMEN

The Dsb protein family is responsible for introducing disulfide bonds into nascent proteins in prokaryotes, stabilizing the structure of many proteins. Helicobacter pylori HP0231 is a Dsb-like protein, shown to catalyze disulfide bond formation and to participate in redox homeostasis. Notably, many H. pylori virulence factors are stabilized by the formation of disulfide bonds. By employing H. pylori HP0231 deficient strains we analyzed the effect of lack of this bacterial protein on the functionality of virulence factors containing putative disulfide bonds. The lack of H. pylori HP0231 impaired CagA translocation into gastric epithelial cells and reduced VacA-induced cellular vacuolation. Moreover, H. pylori HP0231 deficient bacteria were not able to colonize the gastric mucosa of mice, probably due to compromised motility. Together, our data demonstrate an essential function for H. pylori HP0231 in gastric colonization and proper function of bacterial virulence factors related to gastric pathology.


Asunto(s)
Helicobacter pylori/patogenicidad , Estómago/microbiología , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Línea Celular , Helicobacter pylori/metabolismo , Helicobacter pylori/fisiología , Transporte de Proteínas , Vacuolas/microbiología , Virulencia
5.
World J Gastroenterol ; 20(34): 11939-49, 2014 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-25232229

RESUMEN

Helicobacter pylori (H. pylori) is a pathogenic, extracellular bacterium that colonizes the stomach in approximately 50% of the world population. It strongly interacts with the gastric epithelium and mostly causes asymptomatic gastritis. The colonization of H. pylori leads to ulcer development in around 20% of infected patients and may progress to gastric cancer or mucosa-associated lymphoid tissue lymphoma in 1%. Thus, H. pylori is the major cause of gastric cancer worldwide. It has been classified as a class I carcinogen by the World Health Organization. Since its discovery in the early eighties by Warren and Marshall, research has been focused on the investigation of H. pylori biology, host-pathogen interaction, prevention and treatment. Although H. pylori induces a strong humoral and local cellular immune response, the pathogen is not cleared and establishes a chronic infection after encounters in childhood. The ability to colonize the stomach is mediated by several virulence factors that change the host environment, promote adhesion to the epithelium, influence the gastric inflammation and induce immune evasion. H. pylori can be eradicated by antibiotic treatment in combination with a proton-pump inhibitor, but efficacy is decreasing. Current therapies are expensive, have side effects and contribute to increasing antibiotic resistance, underlining the need for novel therapeutics.


Asunto(s)
Vacunas Bacterianas/uso terapéutico , Infecciones por Helicobacter/prevención & control , Helicobacter pylori/patogenicidad , Vacunación , Animales , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/inmunología , Interacciones Huésped-Patógeno , Humanos , Inmunidad Celular , Inmunidad Humoral , Inmunidad Innata , Resultado del Tratamiento , Virulencia
6.
J Immunol ; 193(7): 3566-76, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25172489

RESUMEN

Infection with the gram-negative bacterium Helicobacter pylori is the most prevalent chronic bacterial infection, affecting ∼50% of the world's population, and is the main risk factor of gastric cancer. The proinflammatory cytokine IL-1ß plays a crucial role in the development of gastric tumors and polymorphisms in the IL-1 gene cluster leading to increased IL-1ß production have been associated with increased risk for gastric cancer. To be active, pro-IL-1ß must be cleaved by the inflammasome, an intracellular multiprotein complex implicated in physiological and pathological inflammation. Recently, H. pylori was postulated to activate the inflammasome in murine bone marrow-derived dendritic cells; however, the molecular mechanisms as well as the bacterial virulence factor acting as signal 2 activating the inflammasome remain elusive. In this study, we analyzed the inflammasome complex regulating IL-1ß upon H. pylori infection as well as the molecular mechanisms involved. Our results indicate that H. pylori-induced IL-1ß secretion is mediated by activation of the nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3 inflammasome. We also show that reactive oxygen species, potassium efflux, and lysosomal destabilization are the main cellular mechanisms responsible of nucleotide-binding oligomerization domain family, pyrin domain-containing 3 inflammasome activation upon H. pylori infection, and identify vacuolating cytotoxin A and cag pathogenicity island as the bacterial virulence determinants involved. Moreover, in vivo experiments indicate an important role for the inflammasome in the onset and establishment of H. pylori infection and in the subsequent inflammatory response of the host.


Asunto(s)
Proteínas Bacterianas/inmunología , Proteínas Portadoras/inmunología , Islas Genómicas/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Inmunidad Innata , Inflamasomas/inmunología , Interleucina-1beta/inmunología , Animales , Proteínas Bacterianas/genética , Femenino , Islas Genómicas/genética , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/patología , Helicobacter pylori/genética , Humanos , Masculino , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR
7.
Cytotherapy ; 16(9): 1245-56, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25108651

RESUMEN

BACKGROUND AIMS: Reactivation of cytomegalovirus (CMV) after hematopoietic stem cell transplantation remains a major cause of morbidity despite improved antiviral drug therapies. Selective restoration of CMV immunity by adoptive transfer of CMV-specific T cells is the only alternative approach that has been shown to be effective and non-toxic. We describe the results of clinical-scale isolations of CMV-specific donor lymphocytes with the use of a major histocompatibility (MHC) class I peptide streptamer-based isolation method that yields minimally manipulated cytotoxic T cells of high purity. METHODS: Enrichment of CMV-specific cytotoxic T lymphocytes (CTLs) was performed by labeling 1 × 10(10) leukocytes from a non-mobilized mononuclear cell (MNC) apheresis with MHC class I streptamers and magnetic beads. Thereafter, positively labeled CMV-specific CTLs were isolated through the use of CliniMACS (magnetic-activated cell sorting), and MHC streptamers were released through the use of d-biotin. The purity of enriched CMV-specific CTLs was determined on the basis of MHC streptamer staining and fluorescence-activated cell sorting. RESULTS: A total of 22 processes were performed with the use of five different MHC class I streptamers. The median frequency of CMV-specific CTLs in the starting apheresis product was 0.41% among CD3+ T cells. The isolation process yielded a total of 7.77 × 10(6) CMV-specific CTLs, with a median purity of 90.2%. Selection reagents were effectively removed from the final cell product; the CMV-specific CTLs displayed excellent viability and cytotoxicity and were stable for at least 72 h at 4°C after MNC collection. CONCLUSIONS: Clinical-scale isolation of "minimally manipulated" CMV-specific donor CTLs through the use of MHC class I streptamers is feasible and yields functional CTLs at clinically relevant dosages.


Asunto(s)
Vacunas contra el Cáncer , Separación Celular/métodos , Infecciones por Citomegalovirus/terapia , Citomegalovirus/inmunología , Inmunoterapia Adoptiva/métodos , Linfocitos T Citotóxicos/patología , Antígenos Virales/inmunología , Células Cultivadas , Infecciones por Citomegalovirus/inmunología , Resistencia a Medicamentos , Antígenos de Histocompatibilidad Clase I/química , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Multimerización de Proteína , Recurrencia , Estreptavidina/química , Linfocitos T Citotóxicos/trasplante , Activación Viral
8.
Blood ; 124(4): 628-37, 2014 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-24855206

RESUMEN

Patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) are threatened by potentially lethal viral manifestations like cytomegalovirus (CMV) reactivation. Because the success of today's virostatic treatment is limited by side effects and resistance development, adoptive transfer of virus-specific memory T cells derived from the stem cell donor has been proposed as an alternative therapeutic strategy. In this context, dose minimization of adoptively transferred T cells might be warranted for the avoidance of graft-versus-host disease (GVHD), in particular in prophylactic settings after T-cell-depleting allo-HSCT protocols. To establish a lower limit for successful adoptive T-cell therapy, we conducted low-dose CD8(+) T-cell transfers in the well-established murine Listeria monocytogenes (L.m.) infection model. Major histocompatibility complex-Streptamer-enriched antigen-specific CD62L(hi) but not CD62L(lo) CD8(+) memory T cells proliferated, differentiated, and protected against L.m. infections after prophylactic application. Even progenies derived from a single CD62L(hi) L.m.-specific CD8(+) T cell could be protective against bacterial challenge. In analogy, low-dose transfers of Streptamer-enriched human CMV-specific CD8(+) T cells into allo-HSCT recipients led to strong pathogen-specific T-cell expansion in a compassionate-use setting. In summary, low-dose adoptive T-cell transfer (ACT) could be a promising strategy, particularly for prophylactic treatment of infectious complications after allo-HSCT.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Citomegalovirus/inmunología , Enfermedad Injerto contra Huésped/inmunología , Inmunoterapia Adoptiva , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Inmunodeficiencia Combinada Grave/inmunología , Adolescente , Animales , Diferenciación Celular , Proliferación Celular , Niño , Citomegalovirus/aislamiento & purificación , Infecciones por Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/terapia , Enfermedad Injerto contra Huésped/metabolismo , Enfermedad Injerto contra Huésped/terapia , Trasplante de Células Madre Hematopoyéticas , Proteínas de Homeodominio/fisiología , Humanos , Inmunización , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/fisiología , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Inmunodeficiencia Combinada Grave/metabolismo , Inmunodeficiencia Combinada Grave/terapia , Trasplante Homólogo , Activación Viral
9.
PLoS Pathog ; 9(4): e1003251, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23592983

RESUMEN

Caveolin-1 (Cav1) is a scaffold protein and pathogen receptor in the mucosa of the gastrointestinal tract. Chronic infection of gastric epithelial cells by Helicobacter pylori (H. pylori) is a major risk factor for human gastric cancer (GC) where Cav1 is frequently down-regulated. However, the function of Cav1 in H. pylori infection and pathogenesis of GC remained unknown. We show here that Cav1-deficient mice, infected for 11 months with the CagA-delivery deficient H. pylori strain SS1, developed more severe gastritis and tissue damage, including loss of parietal cells and foveolar hyperplasia, and displayed lower colonisation of the gastric mucosa than wild-type B6129 littermates. Cav1-null mice showed enhanced infiltration of macrophages and B-cells and secretion of chemokines (RANTES) but had reduced levels of CD25+ regulatory T-cells. Cav1-deficient human GC cells (AGS), infected with the CagA-delivery proficient H. pylori strain G27, were more sensitive to CagA-related cytoskeletal stress morphologies ("humming bird") compared to AGS cells stably transfected with Cav1 (AGS/Cav1). Infection of AGS/Cav1 cells triggered the recruitment of p120 RhoGTPase-activating protein/deleted in liver cancer-1 (p120RhoGAP/DLC1) to Cav1 and counteracted CagA-induced cytoskeletal rearrangements. In human GC cell lines (MKN45, N87) and mouse stomach tissue, H. pylori down-regulated endogenous expression of Cav1 independently of CagA. Mechanistically, H. pylori activated sterol-responsive element-binding protein-1 (SREBP1) to repress transcription of the human Cav1 gene from sterol-responsive elements (SREs) in the proximal Cav1 promoter. These data suggested a protective role of Cav1 against H. pylori-induced inflammation and tissue damage. We propose that H. pylori exploits down-regulation of Cav1 to subvert the host's immune response and to promote signalling of its virulence factors in host cells.


Asunto(s)
Caveolina 1/metabolismo , Gastritis/inmunología , Gastritis/microbiología , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/patogenicidad , Animales , Antígenos Bacterianos/genética , Linfocitos B/inmunología , Proteínas Bacterianas/genética , Caveolina 1/deficiencia , Caveolina 1/genética , Línea Celular , Perros , Activación Enzimática , Proteínas Activadoras de GTPasa/metabolismo , Mucosa Gástrica/inmunología , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Gastritis/prevención & control , Células HEK293 , Infecciones por Helicobacter/prevención & control , Helicobacter pylori/genética , Helicobacter pylori/inmunología , Helicobacter pylori/metabolismo , Humanos , Macrófagos/inmunología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Células Parietales Gástricas , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Linfocitos T Reguladores/inmunología , Proteínas Supresoras de Tumor/metabolismo , Proteína Activadora de GTPasa p120/metabolismo
10.
PLoS One ; 7(2): e30543, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22348013

RESUMEN

The confounding consequences of Helicobacter bilis infection in experimental mice populations are well recognized, but the role of this bacterium in human diseases is less known. Limited data are available on virulence determinants of this species. In Helicobacter pylori, γ-glutamyltranspeptidase (γGT) contributes to the colonization of the gastric mucosa and to the pathogenesis of peptic ulcer. The role of γGT in H. bilis infections remains unknown. The annotated genome sequence of H. bilis revealed two putative ggt genes and our aim was to characterize these H. bilis γGT paralogues. We performed a phylogenetic analysis to understand the evolution of Helicobacter γGTs and to predict functional activities of these two genes. In addition, both copies of H. bilis γGTs were expressed as recombinant proteins and their biochemical characteristics were analysed. Functional complementation of Esherichia coli deficient in γGT activity and deletion of γGT in H. bilis were performed. Finally, the inhibitory effect of T-cell and gastric cell proliferation by H. bilis γGT was assessed. Our results indicated that one gene is responsible for γGT activity, while the other showed no γGT activity due to lack of autoprocessing. Although both H. bilis and H. pylori γGTs exhibited a similar affinity to L-Glutamine and γ-Glutamyl-p-nitroanilide, the H. bilis γGT was significantly less active. Nevertheless, H. bilis γGT inhibited T-cell proliferation at a similar level to that observed for H. pylori. Finally, we showed a similar suppressive influence of both H. bilis and H. pylori γGTs on AGS cell proliferation mediated by an apoptosis-independent mechanism. Our data suggest a conserved function of γGT in the Helicobacter genus. Since γGT is present only in a few enterohepatic Helicobacter species, its expression appears not to be essential for colonization of the lower gastrointestinal tract, but it could provide metabolic advantages in colonization capability of different niches.


Asunto(s)
Helicobacter/enzimología , Filogenia , gamma-Glutamiltransferasa/fisiología , Proliferación Celular , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Infecciones por Helicobacter/enzimología , Infecciones por Helicobacter/microbiología , Proteínas Recombinantes , Linfocitos T/microbiología , Linfocitos T/patología , gamma-Glutamiltransferasa/genética
11.
J Immunol ; 184(3): 1617-29, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20042572

RESUMEN

T cells can recognize tumor cells specifically by their TCR and the transfer of TCR-engineered T cells is a promising novel tool in anticancer therapies. We isolated and characterized four allorestricted TCRs with specificity for the HER2/neu-derived peptide 369 (HER2(369)) demonstrating high peptide specificity. PBMCs transduced with especially one TCR, HER2-1, mediated specific tumor reactivity after TCR optimization suggesting that this TCR represents a potential candidate for targeting HER2 by TCR-transduced effector cells. Another TCR showed high-peptide specificity without tumor reactivity. However, the TCR alpha-chain of this TCR specifically recognized HER2(369) not only in combination with the original beta-chain but also with four other beta-chains of the same variable family deriving from TCRs with diverse specificities. Pairing with one beta-chain derived from another HER2(369)-specific TCR potentiated the chimeric TCRs in regard to functional avidity, CD8 independency, and tumor reactivity. Although the frequency of such TCR single chains with dominant peptide recognition is currently unknown, they may represent interesting tools for TCR optimization resulting in enhanced functionality when paired to novel partner chains. However, undirected mispairing with novel partner chains may also result in enhanced cross-reactivity and self-reactivity. These results may have an important impact on the further design of strategies for adoptive transfer using TCR-transduced T cells.


Asunto(s)
Presentación de Antígeno/inmunología , Epítopos de Linfocito T/inmunología , Epítopos Inmunodominantes/metabolismo , Isoantígenos/inmunología , Fragmentos de Péptidos/inmunología , Receptor ErbB-2/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Subgrupos de Linfocitos T/inmunología , Secuencia de Aminoácidos , Línea Celular Transformada , Línea Celular Tumoral , Células Clonales , Epítopos de Linfocito T/metabolismo , Antígeno HLA-A2/inmunología , Antígeno HLA-A2/metabolismo , Humanos , Células Híbridas , Isoantígenos/metabolismo , Células Jurkat , Células K562 , Datos de Secuencia Molecular , Fragmentos de Péptidos/metabolismo , Receptor ErbB-2/metabolismo , Subgrupos de Linfocitos T/metabolismo
12.
Eur J Immunol ; 38(6): 1585-97, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18465771

RESUMEN

Immunization with purified antigens is a safe and practical vaccination strategy but is generally unable to induce sustained CD8(+) T cell-mediated protection against intracellular pathogens. Most efforts to improve the CD8(+) T cell immunogenicity of these vaccines have focused on co-administration of adjuvant to support cross-presentation and dendritic cell maturation. In addition, it has been shown that CD4(+) T cell help during the priming phase contributes to the generation of protective CD8(+) memory T cells. In this report we demonstrate that the depletion of CD4(+) T cells paradoxically enhances long-lasting CD8-mediated protective immunity upon protein vaccination. Functional and genetic in vivo inactivation experiments attribute this enhancement primarily to MHC class II-restricted CD4(+) regulatory T cells (Treg), which appear to physiologically suppress the differentiation process towards long-living effector memory T cells. Since, in functional terms, this suppression by Treg largely exceeds the positive effects of conventional CD4(+) T cell help, even the absence of all CD4(+) T cells or lack of MHC class II-mediated interactions on priming dendritic cells result in enhanced CD8(+) T cell immunogenicity. These findings have important implications for the improvement of vaccines against intracellular pathogens or tumors, especially in patients with highly active Treg.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Reactividad Cruzada/inmunología , Linfocitos T Reguladores/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Antígenos CD4/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Factores de Transcripción Forkhead/genética , Proteínas de Choque Térmico/inmunología , Proteínas Hemolisinas/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Subunidad alfa del Receptor de Interleucina-2/inmunología , Listeriosis/inmunología , Depleción Linfocítica/métodos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Nucleares/genética , Ovalbúmina/inmunología , Fosfoproteínas/inmunología , Bazo/citología , Bazo/inmunología , Bazo/microbiología , Linfocitos T Reguladores/citología , Transactivadores/genética , Vacunación , Proteínas de la Matriz Viral/inmunología
13.
Immunity ; 27(6): 985-97, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18082432

RESUMEN

Upon first antigen encounter, naive CD8(+) T cells get activated, clonally expand, and can develop into very distinct subsets, such as short-living effector cells or different memory subpopulations. The origin of subset diversification is currently unknown, but qualitative and quantitative differences in early signals received by individual precursor cells have been suggested as a major determinant. We show that transfer of a single antigen-specific naive T cell into a normal recipient mouse allowed recovery of clonally expanded daughter cells upon immunization. With this experimental approach, we conclusively demonstrated that a wide range of diversity could develop out of a single precursor cell, including different types of effector and memory T cells. Interestingly, single-cell-derived subset diversification resembled that of polyclonal T cell responses in the same individual mouse, although differentiation patterns differed between immunization strategies. These data implicate that subset diversification is both shaped and synchronized during the expansion phase.


Asunto(s)
Linfocitos T CD8-positivos/citología , Células Madre Hematopoyéticas/citología , Memoria Inmunológica , Subgrupos de Linfocitos T/citología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular , Inmunización , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/fisiología
14.
J Immunol Methods ; 320(1-2): 119-31, 2007 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-17306825

RESUMEN

The development of MHC/peptide multimers has facilitated the visualization and purification of antigen-specific T cells. However, the persistence of multimers leads to prolonged T cell receptor signaling and subsequently to altered T-cell function. We have recently developed a new type of MHC/peptide multimers, which can be dissociated from the T cell. Herein, we have generated and tested for the first time reversible HLA/peptide multimers, termed Streptamers, for the isolation of human T cells. The Streptamer technique demonstrates the specificity and sensitivity of conventional HLA/peptide tetramers with regards to the sorting of human T lymphocytes. This is shown for T cells directed against immunogenic peptides derived from viral and tumor-associated antigens. We show that antigen-specific cytotoxic T cells remain functionally active following Streptamer dissociation, whereas lytic function and proliferation of the T cells is impaired in the presence of conventional tetramers. These novel HLA/peptide Streptamer reagents allow the isolation of antigen-specific T cells with preserved function and, therefore, facilitate the development of adoptive T cell transfer regimens for the treatment of patients with cancer or infectious diseases.


Asunto(s)
Antígenos de Neoplasias/inmunología , Antígenos Virales/inmunología , Antígenos HLA/aislamiento & purificación , Linfocitos T Citotóxicos/inmunología , Células Cultivadas , Citomegalovirus/inmunología , Antígenos HLA/química , Antígenos HLA/inmunología , Humanos , Antígeno MART-1 , Proteínas de Neoplasias/inmunología , Linfocitos T Citotóxicos/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...