Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 600(7890): 701-706, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34673755

RESUMEN

Following severe adverse reactions to the AstraZeneca ChAdOx1-S-nCoV-19 vaccine1,2, European health authorities recommended that patients under the age of 55 years who received one dose of ChAdOx1-S-nCoV-19 receive a second dose of the Pfizer BNT162b2 vaccine as a booster. However, the effectiveness and the immunogenicity of this vaccination regimen have not been formally tested. Here we show that the heterologous ChAdOx1-S-nCoV-19 and BNT162b2 combination confers better protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection than the homologous BNT162b2 and BNT162b2 combination in a real-world observational study of healthcare workers (n = 13,121). To understand the underlying mechanism, we conducted a longitudinal survey of the anti-spike immunity conferred by each vaccine combination. Both combinations induced strong anti-spike antibody responses, but sera from heterologous vaccinated individuals displayed a stronger neutralizing activity regardless of the SARS-CoV-2 variant. This enhanced neutralizing potential correlated with increased frequencies of switched and activated memory B cells that recognize the SARS-CoV-2 receptor binding domain. The ChAdOx1-S-nCoV-19 vaccine induced a weaker IgG response but a stronger T cell response than the BNT162b2 vaccine after the priming dose, which could explain the complementarity of both vaccines when used in combination. The heterologous vaccination regimen could therefore be particularly suitable for immunocompromised individuals.


Asunto(s)
Vacuna BNT162/administración & dosificación , Vacuna BNT162/inmunología , COVID-19/inmunología , COVID-19/prevención & control , ChAdOx1 nCoV-19/administración & dosificación , ChAdOx1 nCoV-19/inmunología , SARS-CoV-2/inmunología , Vacunación/estadística & datos numéricos , Adulto , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Femenino , Francia/epidemiología , Hospitales Universitarios , Humanos , Memoria Inmunológica/inmunología , Incidencia , Masculino , Células B de Memoria/inmunología , Células T de Memoria/inmunología , Persona de Mediana Edad , Glicoproteína de la Espiga del Coronavirus/inmunología
2.
Eur J Immunol ; 54(7): e2451035, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38627984

RESUMEN

OBJECTIVES: In the post-SARS-CoV-2 pandemic era, "breakthrough infections" are still documented, due to variants of concerns (VoCs) emergence and waning humoral immunity. Despite widespread utilization, the definition of the anti-Spike (S) immunoglobulin-G (IgG) threshold to define protection has unveiled several limitations. Here, we explore the advantages of incorporating T-cell response assessment to enhance the definition of immune memory profile. METHODS: SARS-CoV-2 interferon-gamma release assay test (IGRA) was performed on samples collected longitudinally from immunocompetent healthcare workers throughout their immunization by infection and/or vaccination, anti-receptor-binding domain IgG levels were assessed in parallel. The risk of symptomatic infection according to cellular/humoral immune capacities during Omicron BA.1 wave was then estimated. RESULTS: Close to 40% of our samples were exclusively IGRA-positive, largely due to time elapsed since their last immunization. This suggests that individuals have sustained long-lasting cellular immunity, while they would have been classified as lacking protective immunity based solely on IgG threshold. Moreover, the Cox regression model highlighted that Omicron BA.1 circulation raises the risk of symptomatic infection while increased anti-receptor-binding domain IgG and IGRA levels tended to reduce it. CONCLUSION: The discrepancy between humoral and cellular responses highlights the significance of assessing the overall adaptive immune response. This integrated approach allows the identification of vulnerable subjects and can be of interest to guide antiviral prophylaxis at an individual level.


Asunto(s)
Anticuerpos Antivirales , COVID-19 , Inmunidad Humoral , Inmunoglobulina G , Memoria Inmunológica , Ensayos de Liberación de Interferón gamma , SARS-CoV-2 , Humanos , COVID-19/inmunología , SARS-CoV-2/inmunología , Memoria Inmunológica/inmunología , Inmunidad Humoral/inmunología , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Inmunoglobulina G/inmunología , Inmunoglobulina G/sangre , Masculino , Femenino , Adulto , Persona de Mediana Edad , Ensayos de Liberación de Interferón gamma/métodos , Glicoproteína de la Espiga del Coronavirus/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Linfocitos T/inmunología , Personal de Salud , Vacunas contra la COVID-19/inmunología
3.
J Allergy Clin Immunol ; 150(5): 1194-1208, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35779666

RESUMEN

BACKGROUND: Epicutaneous immunotherapy (EPIT) protocols have recently been developed to restore tolerance in patients with food allergy. The mechanisms by which EPIT protocols promote desensitization rely on a profound immune deviation of pathogenic T- and B-cell responses. OBJECTIVE: To date, little is known about the contribution of skin dendritic cells (skDCs) to T-cell remodeling and EPIT efficacy. METHODS: We capitalized on a preclinical model of food allergy to ovalbumin (OVA) to characterize the phenotype and functions of OVA+ skDCs throughout the course of EPIT. RESULTS: Our results showed that both Langerhans cells and dermal conventional cDC1 and cDC2 subsets retained their ability to capture OVA in the skin and to migrate toward the skin-draining lymph nodes during EPIT. However, their activation/maturation status was significantly impaired, as evidenced by the gradual and selective reduction of CD86, CD40, and OVA protein expression in respective subsets. Phenotypic changes during EPIT were also characterized by a progressive diversification of single-cell gene signatures within each DC subset. Interestingly, we observed that OVA+ Langerhans cells progressively lost their capacity to prime CD4+ TEFF cells, but gained regulatory T-cell stimulatory properties. In contrast, cDC1 were inefficient in priming CD4+ TEFF cells or in reactivating TMEM cells in vitro, whereas cDC2 retained moderate stimulatory properties, and progressively biased type 2 immunity toward type 1 and type 17 responses. CONCLUSIONS: Our results therefore emphasize that the acquisition of distinct phenotypic and functional specializations by skDCs during EPIT is at the cornerstone of the desensitization process.


Asunto(s)
Hipersensibilidad a los Alimentos , Células de Langerhans , Humanos , Desensibilización Inmunológica/métodos , Ovalbúmina , Linfocitos T Reguladores , Alérgenos
5.
In Silico Biol ; 14(1-2): 13-39, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33554899

RESUMEN

To develop vaccines it is mandatory yet challenging to account for inter-individual variability during immune responses. Even in laboratory mice, T cell responses of single individuals exhibit a high heterogeneity that may come from genetic backgrounds, intra-specific processes (e.g. antigen-processing and presentation) and immunization protocols.To account for inter-individual variability in CD8 T cell responses in mice, we propose a dynamical model coupled to a statistical, nonlinear mixed effects model. Average and individual dynamics during a CD8 T cell response are characterized in different immunization contexts (vaccinia virus and tumor). On one hand, we identify biological processes that generate inter-individual variability (activation rate of naive cells, the mortality rate of effector cells, and dynamics of the immunogen). On the other hand, introducing categorical covariates to analyze two different immunization regimens, we highlight the steps of the response impacted by immunogens (priming, differentiation of naive cells, expansion of effector cells and generation of memory cells). The robustness of the model is assessed by confrontation to new experimental data.Our approach allows to investigate immune responses in various immunization contexts, when measurements are scarce or missing, and contributes to a better understanding of inter-individual variability in CD8 T cell immune responses.


Asunto(s)
Linfocitos T CD8-positivos , Virus Vaccinia , Animales , Antígenos , Inmunización , Ratones , Vacunación
6.
Proc Natl Acad Sci U S A ; 115(41): 10404-10409, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30249647

RESUMEN

Prominent changes in the gut microbiota (referred to as "dysbiosis") play a key role in the development of allergic disorders, but the underlying mechanisms remain unknown. Study of the delayed-type hypersensitivity (DTH) response in mice contributed to our knowledge of the pathophysiology of human allergic contact dermatitis. Here we report a negative regulatory role of the RIG-I-like receptor adaptor mitochondrial antiviral signaling (MAVS) on DTH by modulating gut bacterial ecology. Cohousing and fecal transplantation experiments revealed that the dysbiotic microbiota of Mavs-/- mice conferred a proallergic phenotype that is communicable to wild-type mice. DTH sensitization coincided with increased intestinal permeability and bacterial translocation within lymphoid organs that enhanced DTH severity. Collectively, we unveiled an unexpected impact of RIG-I-like signaling on the gut microbiota with consequences on allergic skin disease outcome. Primarily, these data indicate that manipulating the gut microbiota may help in the development of therapeutic strategies for the treatment of human allergic skin pathologies.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Disbiosis/complicaciones , Microbioma Gastrointestinal/inmunología , Hipersensibilidad/etiología , Intestinos/inmunología , Enfermedades Cutáneas Bacterianas/etiología , Animales , Modelos Animales de Enfermedad , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Hipersensibilidad/metabolismo , Hipersensibilidad/patología , Intestinos/microbiología , Intestinos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Transducción de Señal , Enfermedades Cutáneas Bacterianas/metabolismo , Enfermedades Cutáneas Bacterianas/patología
7.
J Immunol ; 200(2): 551-557, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29237776

RESUMEN

The role of sphingosine-1 phosphate (S1P) in leukocyte trafficking has been well deciphered in mice but remains largely unaddressed in humans. In this study, we assessed the ex vivo response to S1P of primary human T cell subsets. We found that tonsil but not blood leukocytes were responsive to S1P gradients, suggesting that T cell responsiveness is regulated during their recirculation in vivo. Tonsil naive T cells were readily chemoattracted by S1P in an FTY720-sensitive, S1PR1-dependent manner. Surprisingly, S1P had the opposite effect on effector memory T cells, resident memory T cells, and recently activated T cells, inhibiting their spontaneous or chemokine-induced migration. This inhibition was also more pronounced for CD4 T cells than for CD8 T cell subsets, and was dependent on S1PR2, as shown using the S1PR2 antagonist JTE-013. S1PR1 was progressively downregulated during T cell differentiation whereas S1PR2 expression remained stable. Our results suggest that the ratio between S1PR1 and S1PR2 governs the migratory behavior of T cell subsets. They also challenge previous models of the role of S1P in lymphocyte recirculation and suggest that S1P promotes retention of memory T cell subsets in secondary lymphoid organs, via S1PR2.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Memoria Inmunológica , Lisofosfolípidos/inmunología , Esfingosina/análogos & derivados , Linfocitos T/inmunología , Quimiocinas/metabolismo , Humanos , Inmunofenotipificación , Activación de Linfocitos/inmunología , Tejido Linfoide/inmunología , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Esfingosina/inmunología , Receptores de Esfingosina-1-Fosfato , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T/metabolismo
8.
J Immunol ; 200(10): 3635-3646, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29632146

RESUMEN

The pool of memory-phenotype CD8 T cells is composed of Ag-induced (AI) and cytokine-induced innate (IN) cells. IN cells have been described as having properties similar to those of AI memory cells. However, we found that pathogen-induced AI memory cells can be distinguished in mice from naturally generated IN memory cells by surface expression of NKG2D. Using this marker, we described the increased functionalities of AI and IN memory CD8 T cells compared with naive cells, as shown by comprehensive analysis of cytokine secretion and gene expression. However, AI differed from IN memory CD8 T cells by their capacity to migrate to the lung parenchyma upon inflammation or infection, a process dependent on their expression of ITGA1/CD49a and ITGA4/CD49d integrins.


Asunto(s)
Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Pulmón/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Virosis/inmunología , Animales , Citocinas/inmunología , Inflamación/inmunología , Inflamación/virología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
9.
Biomacromolecules ; 18(12): 4022-4033, 2017 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-29020442

RESUMEN

One of the challenges of photodynamic therapy is to increase the penetration depth of light irradiation in the tumor tissues. Although two-photon excitation strategies have been developed, the two-photon absorption cross sections of clinically used photosensitizers are generally low (below 300 GM). Besides, photosensitizers with high cross section values are often non-water-soluble. In this research work, a whole family of photosensitizer-polymer conjugates was synthesized via the covalent binding of a photosensitizer with a relatively high cross section along a biocompatible copolymer chain. The resulting photosensitizer-polymer conjugates were water-soluble and could be imaged in cellulo by two-photon microscopy thanks to their high two-photon absorption cross sections (up to 2600 GM in water, in the NIR range). In order to explore the structure/photodynamic activity relationship of such macromolecular photosensitizers, the influence of the polymer size, photosensitizer density, and presence of charges along the polymer backbone was investigated (neutral, anionic, cationic, and zwitterionic conjugates were compared). The macromolecular photosensitizers were not cytotoxic in the absence of light irradiation. Their kinetics of cellular uptake in the B16-F10 melanoma cell line were followed by flow cytometry over 24 h. The efficiency of cell death upon photoactivation was found to be highly correlated to the cellular uptake in turn correlated to the global charge of the macromolecular photosensitizer which appeared as the determining structural parameter.


Asunto(s)
Muerte Celular/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Polímeros/química , Polímeros/farmacología , Animales , Línea Celular Tumoral , Fluorescencia , Sustancias Macromoleculares/farmacología , Melanoma/tratamiento farmacológico , Ratones , Estructura Molecular , Tamaño de la Partícula , Fotoquimioterapia/métodos , Fotones , Relación Estructura-Actividad
10.
J Infect Dis ; 211(4): 571-81, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25006047

RESUMEN

BACKGROUND: Bone and joint infection, mainly caused by Staphylococcus aureus, is associated with significant morbidity and mortality, characterized by severe inflammation and progressive bone destruction. Studies mostly focused on the interaction between S. aureus and osteoblasts, the bone matrix-forming cells, while interactions between S. aureus and osteoclasts, the only cells known to be able to degrade bone, have been poorly explored. METHODS: We developed an in vitro infection model of primary murine osteoclasts to study the direct impact of live S. aureus on osteoclastogenesis and osteoclast resorption activity. RESULTS: Staphylococcal infection of bone marrow-derived osteoclast precursors induced their differentiation into activated macrophages that actively secreted proinflammatory cytokines. These cytokines enhanced the bone resorption capacity of uninfected mature osteoclasts and promoted osteoclastogenesis of the uninfected precursors at the site of infection. Moreover, infection of mature osteoclasts by live S. aureus directly enhanced their ability to resorb bone by promoting cellular fusion. CONCLUSIONS: Our results highlighted two complementary mechanisms involved in bone loss during bone and joint infection, suggesting that osteoclasts could be a pivotal target for limiting bone destruction.


Asunto(s)
Resorción Ósea/microbiología , Interacciones Huésped-Patógeno/fisiología , Osteoclastos/microbiología , Osteoclastos/fisiología , Staphylococcus aureus/patogenicidad , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Citocinas/metabolismo , Durapatita , Ratones , Modelos Biológicos , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética
11.
Int J Cancer ; 136(5): 1085-94, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25046660

RESUMEN

Dendritic cells (DCs) cross-present antigen (Ag) to initiate T-cell immunity against most infections and tumors. Natural killer (NK) cells are innate cytolytic lymphocytes that have emerged as key modulators of multiple DC functions. Here, we show that human NK cells promote cross-presentation of tumor cell-derived Ag by DC leading to Ag-specific CD8(+) T-cell activation. Surprisingly, cytotoxic function of NK cells was not required. Instead, we highlight a critical and nonredundant role for IFN-γ and TNF-α production by NK cells to enhance cross-presentation by DC using two different Ag models. Importantly, we observed that NK cells promote cell-associated Ag cross-presentation selectively by monocytes-derived DC (Mo-DC) and CD34-derived CD11b(neg) CD141(high) DC subsets but not by myeloid CD11b(+) DC. Moreover, we demonstrate that triggering NK cell activation by monoclonal antibodies (mAbs)-coated tumor cells leads to efficient DC cross-presentation, supporting the concept that NK cells can contribute to therapeutic mAbs efficiency by inducing downstream adaptive immunity. Taken together, our findings point toward a novel role of human NK cells bridging innate and adaptive immunity through selective induction of cell-associated Ag cross-presentation by CD141(high) DC, a process that could be exploited to better harness Ag-specific cellular immunity in immunotherapy.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos de Neoplasias/inmunología , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Inmunidad Celular/inmunología , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Células Dendríticas/patología , Humanos , Células Asesinas Naturales/patología , Neoplasias/patología , Células Tumorales Cultivadas
12.
Trends Immunol ; 33(8): 381-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22677187

RESUMEN

Cross-presentation by dendritic cells (DCs) of exogenous antigens on MHC class I is important for the generation of immune responses to intracellular pathogens, as well as for maintenance of self tolerance. In mice, the CD8(+) DC lineage is specialised for this role. However, DCs of this lineage are not born with cross-presentation capacity. Several studies have demonstrated that it must be induced as a later developmental step by cytokines such as granulocyte macrophage colony-stimulating factor (GM-CSF), or by microbial products such as toll-like receptor (TLR) ligands. Increased cross-presentation capacity is thus induced in peripheral CD8 lineage DCs during inflammation or infection. However, this capacity is already fully developed in steady-state thymic CD8(+) DCs, in accordance with their role in the deletion of self-reactive developing T cells.


Asunto(s)
Antígenos/inmunología , Reactividad Cruzada , Células Dendríticas/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linaje de la Célula , Células Dendríticas/citología , Humanos
13.
J Immunol ; 191(7): 3847-57, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23975862

RESUMEN

The inflammasome is a signaling platform that is central to the innate immune responses to bacterial infections. Francisella tularensis is a bacterium replicating within the host cytosol. During F. tularensis subspecies novicida infection, AIM2, an inflammasome receptor sensing cytosolic DNA, activates caspase-1 in an ASC-dependent manner, leading to both pyroptosis and release of the proinflammatory cytokines IL-1ß and IL-18. Activation of this canonical inflammasome pathway is key to limit F. novicida infection. In this study, by comparing the immune responses of AIM2 knockout (KO), ASC(KO), and Casp1(KO) mice in response to F. novicida infection, we observed that IFN-γ levels in the serum of Casp1(KO) mice were much higher than the levels observed in AIM2(KO) and ASC(KO) mice. This difference in IFN-γ production was due to a large production of IFN-γ by NK cells in Casp1(KO) mice that was not observed in ASC(KO) mice. The deficit in IFN-γ production observed in ASC(KO) mice was not due to a reduced Dock2 expression or to an intrinsic defect of ASC(KO) NK cells. We demonstrate that in infected Casp1(KO) mice, IFN-γ production is due to an ASC-dependent caspase-1-independent pathway generating IL-18. Furthermore, we present in vitro data suggesting that the recently described AIM2/ASC/caspase-8 noncanonical pathway is responsible for the caspase-1-independent IL-18 releasing activity. To our knowledge, this study is the first in vivo evidence of an alternative pathway able to generate in a caspase-1-independent pathway bioactive IL-18 to boost the production of IFN-γ, a cytokine critical for the host antibacterial response.


Asunto(s)
Caspasa 1/deficiencia , Proteínas del Citoesqueleto/genética , Francisella , Infecciones por Bacterias Gramnegativas/genética , Infecciones por Bacterias Gramnegativas/metabolismo , Interferón gamma/metabolismo , Interleucina-18/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Adaptadoras de Señalización CARD , Caspasa 1/genética , Caspasa 8/genética , Caspasa 8/metabolismo , Degranulación de la Célula/inmunología , Diferenciación Celular/inmunología , Proteínas del Citoesqueleto/metabolismo , Modelos Animales de Enfermedad , Francisella/inmunología , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Infecciones por Bacterias Gramnegativas/inmunología , Factores de Intercambio de Guanina Nucleótido , Células HEK293 , Humanos , Inflamasomas/metabolismo , Interleucina-18/sangre , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Ratones , Ratones Noqueados
14.
Eur J Immunol ; 43(6): 1667-75, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23519784

RESUMEN

Patrolling Ly6C(-) monocytes are blood-circulating cells that play a role in inflammation and in the defense against pathogens. Here, we show that similar to natural killer (NK) cells, patrolling monocytes express high levels of S1PR5, a G-coupled receptor for sphingosine-1 phosphate. We found that S1pr5(-/-) mice lack peripheral Ly6C(-) monocytes but have a normal number of these cells in the bone marrow (BM). Various lines of evidence exclude a direct contribution of S1PR5 in the survival of Ly6C(-) monocytes at the periphery. Rather, our data support a role for S1PR5 in the egress of Ly6C(-) monocytes from the BM. In particular, we observed a reduced frequency of patrolling monocytes in BM sinusoids of S1PR5 KO mice. Unexpectedly, S1P was not a chemoattractant for patrolling monocytes and had no significant effect on their viability in vitro. Moreover, the disruption of S1P gradients in vivo did not alter Ly6C(-) monocyte trafficking and viability. These data suggest that S1PR5 regulates the trafficking of monocytes via a mechanism independent of S1P gradients.


Asunto(s)
Antígenos Ly/metabolismo , Médula Ósea/inmunología , Monocitos/inmunología , Receptores de Lisoesfingolípidos/metabolismo , Animales , Circulación Sanguínea , Movimiento Celular/inmunología , Supervivencia Celular , Células Cultivadas , Femenino , Homeostasis , Vigilancia Inmunológica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Lisoesfingolípidos/genética
15.
J Immunol ; 189(7): 3480-9, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22942430

RESUMEN

IL-4 is one of the main cytokines produced during Th2-inducing pathologies. This cytokine has been shown to affect a number of immune processes such as Th differentiation and innate immune responses. However, the impact of IL-4 on CD8 T cell responses remains unclear. In this study, we analyzed the effects of IL-4 on global gene expression profiles of Ag-induced memory CD8 T cells in the mouse. Gene ontology analysis of this signature revealed that IL-4 regulated most importantly genes associated with immune responses. Moreover, this IL-4 signature overlapped with the set of genes preferentially expressed by memory CD8 T cells over naive CD8 T cells. In particular, IL-4 downregulated in vitro and in vivo in a STAT6-dependent manner the memory-specific expression of NKG2D, thereby increasing the activation threshold of memory CD8 T cells. Furthermore, IL-4 impaired activation of memory cells as well as their differentiation into effector cells. This phenomenon could have an important clinical relevance as patients affected by Th2 pathologies such as parasitic infections or atopic dermatitis often suffer from viral-induced complications possibly linked to inefficient CD8 T cell responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Regulación hacia Abajo/inmunología , Memoria Inmunológica , Interleucina-4/fisiología , Subfamilia K de Receptores Similares a Lectina de Células NK/antagonistas & inhibidores , Subfamilia K de Receptores Similares a Lectina de Células NK/biosíntesis , Animales , Células Cultivadas , Quimiocina CCL5/antagonistas & inhibidores , Quimiocina CCL5/metabolismo , Regulación hacia Abajo/genética , Humanos , Inmunidad Innata/genética , Memoria Inmunológica/genética , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Factor de Transcripción STAT6/fisiología
16.
Vaccines (Basel) ; 12(3)2024 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-38543950

RESUMEN

Dengue virus (DENV) infection continues to be a public health challenge, lacking a specific cure. Vaccination remains the primary strategy against dengue; however, existing live-attenuated vaccines display variable efficacy across four serotypes, influenced by host serostatus and age, and predominantly inducing humoral responses. To address this limitation, this study investigates a multiepitope-based immunogen designed to induce robust cellular immunity across all DENV serotypes. The chimeric immunogen integrates H-2d specific MHC-I binding T-cell epitopes derived from conserved domains within the DENV envelope protein. Immuno-informatics analyses supported its stability, non-allergenic nature, and strong MHC-I binding affinity as an antigen. To assess the immunogenicity of the multiepitope, it was expressed in murine bone-marrow-derived dendritic cells (BMDCs) that were used to prime mice. In this experimental model, simultaneous exposure to T-cell epitopes from all four DENV serotypes initiated distinct IFNγ-CD8 T-cell responses for different serotypes. These results supported the potential of the multiepitope construct as a vaccine candidate. While the optimization of the immunogen design remains a continuous pursuit, this proof-of-concept study provides a starting point for evaluating its protective efficacy against dengue infection in vivo. Moreover, our results support the development of a multiepitope vaccine that could trigger a pan-serotype anti-dengue CD8 response.

17.
iScience ; 27(4): 109411, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38510150

RESUMEN

To investigate the impact of paracrine IL-2 signals on memory precursor (MP) cell differentiation, we activated CD8 T cell in vitro in the presence or absence of exogenous IL-2 (ex-IL-2). We assessed memory differentiation by transferring these cells into virus-infected mice. Both conditions generated CD8 T cells that participate in the ongoing response and gave rise to similar memory cells. Nevertheless, when transferred into a naive host, T cells activated with ex-IL-2 generated a higher frequency of memory cells displaying increased functional memory traits. Single-cell RNA-seq analysis indicated that without ex-IL-2, cells rapidly acquire an MP signature, while in its presence they adopted an effector signature. This was confirmed at the protein level and in a functional assay. Overall, ex-IL-2 delays the transition into MP cells, allowing the acquisition of effector functions that become imprinted in their progeny. These findings may help to optimize the generation of therapeutic T cells.

18.
Genesis ; 51(3): 193-200, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23281269

RESUMEN

Immune tolerance to self-antigens is a complex process that utilizes multiple mechanisms working in concert to maintain homeostasis and prevent autoimmunity. Considerable progress in deciphering the mechanisms controlling the activation or deletion of T cells has been made by using T cell receptor (TCR) transgenic mice. One such model is the F5 model in which CD8 T cells express a TCR specific for an epitope derived from the influenza NP68 protein. Our aim was to create transgenic mouse models expressing constitutively the NP68 epitope fused to enhanced green fluorescent protein (EGFP) in order to assess unambiguously the relative levels of NP68 epitope expressed by single cells. We used a lentiviral-based approach to generate two independent transgenic mouse strains expressing the fusion protein EGFP-NP68 under the control of CAG (CMV immediate early enhancer and the chicken ß-actin promoter) or spleen focus-forming virus (SFFV) promoters. Analysis of the pattern of EGFP expression in the hematopoietic compartment showed that CAG and SFFV promoters are differentially regulated during T cell development. However, both promoters drove high EGFP-NP68 expression in dendritic cells (pDCs, CD8α(+) cDCs, and CD8α(-) cDCs) from spleen or generated in vitro following differentiation from bone-marrow progenitors. NP68 epitope was properly processed and successfully presented by dendritic cells (DCs) by direct presentation and cross-presentation to F5 CD8 T cells. The models presented here are valuable tools to investigate the priming of F5 CD8 T cells by different subsets of DCs.


Asunto(s)
Epítopos de Linfocito T/genética , Proteínas Fluorescentes Verdes/genética , Transgenes , Proteínas Virales/genética , Animales , Células Dendríticas/metabolismo , Ingeniería Genética/métodos , Vectores Genéticos , Antígenos de Histocompatibilidad Clase I/metabolismo , Lentivirus/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas , Linfocitos T/metabolismo
19.
Blood ; 118(18): 4863-71, 2011 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-21911833

RESUMEN

During development, natural killer (NK) cells exit the BM to reach the blood. CXCR4 retains NK cells in the BM, whereas the sphingosine-1 phosphate receptor 5 (S1P5) promotes their exit from this organ. However, how the action of these receptors is coordinated to preserve NK-cell development in the BM parenchyma while providing mature NK cells at the periphery is unclear. The role of CXCR4 and S1P5 in NK-cell recirculation at the periphery is also unknown. In the present study, we show that, during NK-cell differentiation, CXCR4 expression decreases whereas S1P5 expression increases, thus favoring the exit of mature NK cells via BM sinusoids. Using S1P5(-/-) mice and a new knockin mouse model in which CXCR4 cannot be desensitized (a mouse model of warts, hypogammaglobulinemia, infections, and myelokathexis [WHIM] syndrome), we demonstrate that NK-cell exit from the BM requires both CXCR4 desensitization and S1P5 engagement. These 2 signals occur independently of each other: CXCR4 desensitization is not induced by S1P5 engagement and vice versa. Once in the blood, the S1P concentration increases and S1P5 responsiveness decreases. This responsiveness is recovered in the lymph nodes to allow NK-cell exit via lymphatics in a CXCR4-independent manner. Therefore, coordinated changes in CXCR4 and S1P5 responsiveness govern NK-cell trafficking.


Asunto(s)
Movimiento Celular , Desensibilización Inmunológica , Células Asesinas Naturales/fisiología , Receptores CXCR4/inmunología , Receptores de Lisoesfingolípidos/inmunología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/fisiología , Movimiento Celular/genética , Movimiento Celular/inmunología , Movimiento Celular/fisiología , Desensibilización Inmunológica/métodos , Técnicas de Sustitución del Gen , Esquemas de Inmunización , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores CXCR4/genética , Receptores CXCR4/fisiología , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/fisiología
20.
J Immunol ; 186(3): 1503-11, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21187449

RESUMEN

Cross-presentation of cell-associated Ags by dendritic cells (DC) plays an important role in immunity. DC in lymphoid tissues are short lived, being continuously replaced by precursors that proliferate and differentiate locally. Paradoxically, although TLR ligands promote immune responses and stimulate DC replenishment, they impair the cross-priming capacity of terminally differentiated splenic CD8α(+) DC, the major subset involved in cross-priming. In this study, we have investigated the cross-presentation capacity of newly generated murine DC and especially immediate precursors of CD8α(+) DC. We show that these DC do not cross-present Ag from dead cells unless stimulated by TLR ligands before Ag capture. TLR ligand CpG induced the expression of costimulatory molecules required for CD8 T cell activation but also regulated the intracellular mechanisms of cross-presentation such as Ag degradation rates without regulating Ag uptake. GM-CSF, an inflammatory cytokine associated with infections, also promoted cross-presentation acquisition by pre-CD8α(+) DC and synergized with TLR9 ligand. The concept that TLR ligands as well as inflammatory cytokines promote the acquisition of cross-presenting properties by pre-CD8α(+) DC has important implications during immune responses and when considering the use of these cells for vaccination.


Asunto(s)
Antígenos/metabolismo , Células de la Médula Ósea/inmunología , Antígenos CD8/biosíntesis , Islas de CpG/fisiología , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Proteínas de la Membrana/metabolismo , Células Madre/inmunología , Animales , Antígenos/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Antígenos CD8/inmunología , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Línea Celular Tumoral , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/patología , Ligandos , Melanoma Experimental , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Necrosis , Células Madre/metabolismo , Células Madre/patología , Receptores Toll-Like/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA