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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 19(4): 386-396, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29556002

RESUMEN

Live attenuated vaccines are generally highly efficacious and often superior to inactivated vaccines, yet the underlying mechanisms of this remain largely unclear. Here we identify recognition of microbial viability as a potent stimulus for follicular helper T cell (TFH cell) differentiation and vaccine responses. Antigen-presenting cells (APCs) distinguished viable bacteria from dead bacteria through Toll-like receptor 8 (TLR8)-dependent detection of bacterial RNA. In contrast to dead bacteria and other TLR ligands, live bacteria, bacterial RNA and synthetic TLR8 agonists induced a specific cytokine profile in human and porcine APCs, thereby promoting TFH cell differentiation. In domestic pigs, immunization with a live bacterial vaccine induced robust TFH cell and antibody responses, but immunization with its heat-killed counterpart did not. Finally, a hypermorphic TLR8 polymorphism was associated with protective immunity elicited by vaccination with bacillus Calmette-Guérin (BCG) in a human cohort. We have thus identified TLR8 as an important driver of TFH cell differentiation and a promising target for TFH cell-skewing vaccine adjuvants.


Asunto(s)
Activación de Linfocitos/inmunología , Viabilidad Microbiana/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Receptor Toll-Like 8/inmunología , Vacunas Atenuadas/inmunología , Adulto , Animales , Formación de Anticuerpos/inmunología , Diferenciación Celular/inmunología , Femenino , Humanos , Masculino , Porcinos
2.
Immunity ; 49(1): 120-133.e9, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-30005826

RESUMEN

B lymphocytes can suppress immunity through interleukin (IL)-10 production in infectious, autoimmune, and malignant diseases. Here, we have identified a natural plasma cell subset that distinctively expresses the inhibitory receptor LAG-3 and mediates this function in vivo. These plasma cells also express the inhibitory receptors CD200, PD-L1, and PD-L2. They develop from various B cell subsets in a B cell receptor (BCR)-dependent manner independently of microbiota in naive mice. After challenge they upregulate IL-10 expression via a Toll-like receptor-driven mechanism within hours and without proliferating. This function is associated with a unique transcriptome and epigenome, including the lowest amount of DNA methylation at the Il10 locus compared to other B cell subsets. Their augmented accumulation in naive mutant mice with increased BCR signaling correlates with the inhibition of memory T cell formation and vaccine efficacy after challenge. These natural regulatory plasma cells may be of broad relevance for disease intervention.


Asunto(s)
Antígenos CD/genética , Expresión Génica , Interleucina-10/biosíntesis , Células Plasmáticas/inmunología , Animales , Antígenos CD/inmunología , Subgrupos de Linfocitos B/inmunología , Epigénesis Genética , Femenino , Perfilación de la Expresión Génica , Interleucina-10/genética , Activación de Linfocitos , Masculino , Ratones , Células Plasmáticas/fisiología , Receptores de Antígenos de Linfocitos B/metabolismo , Salmonelosis Animal/inmunología , Transducción de Señal , Linfocitos T/inmunología , Receptores Toll-Like/metabolismo , Regulación hacia Arriba/genética , Vacunas/inmunología , Proteína del Gen 3 de Activación de Linfocitos
3.
Am J Respir Crit Care Med ; 204(12): 1403-1417, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34534436

RESUMEN

Rationale: Pulmonary sarcoidosis is generally presumed to be a T-helper cell type 1- and macrophage-driven disease. However, mouse models have recently revealed that chronically inflamed lung tissue can also comprise T follicular helper (Tfh)-like cells and represents a site of active T-cell/B-cell cooperation. Objectives: To assess the role of pulmonary Tfh- and germinal center-like lymphocytes in sarcoidosis. Methods: BAL fluid, lung tissue, and peripheral blood samples from patients with sarcoidosis were analyzed by flow cytometry, immunohistology, RNA sequencing, and in vitro T-cell/B-cell cooperation assays for phenotypic and functional characterization of germinal center-like reactions in inflamed tissue. Measurements and Main Results: We identified a novel population of Tfh-like cells characterized by high expression of the B helper molecules CD40L and IL-21 in BAL of patients with sarcoidosis. Transcriptome analysis further confirmed a phenotype that was both Tfh-like and tissue resident. BAL T cells provided potent help for B cells to differentiate into antibody-producing cells. In lung tissue, we observed large peribronchial infiltrates with T and B cells in close contact, and many IgA+ plasmablasts. Most clusters were nonectopic; that is, they did not contain follicular dendritic cells. Patients with sarcoidosis also showed elevated levels of PD-1high CXCR5- CD40Lhigh ICOShigh Tfh-like cells, but not classical CXCR5+ Tfh cells, in the blood. Conclusions: Active T-cell/B-cell cooperation and local production of potentially pathogenic antibodies in the inflamed lung represents a novel pathomechanism in sarcoidosis and should be considered from both diagnostic and therapeutic perspectives.


Asunto(s)
Linfocitos B/inmunología , Centro Germinal/inmunología , Pulmón/inmunología , Sarcoidosis Pulmonar/inmunología , Células T Auxiliares Foliculares/inmunología , Adulto , Anciano , Biomarcadores/sangre , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Estudios de Casos y Controles , Femenino , Citometría de Flujo , Humanos , Pulmón/patología , Masculino , Persona de Mediana Edad , Sarcoidosis Pulmonar/sangre , Sarcoidosis Pulmonar/patología
4.
Immunology ; 163(1): 86-97, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33427298

RESUMEN

IL-33 is a member of the IL-1 family. By binding to its receptor ST2 (IL-33R) on mast cells, IL-33 induces the MyD88-dependent activation of the TAK1-IKK2 signalling module resulting in activation of the MAP kinases p38, JNK1/2 and ERK1/2, and of NFκB. Depending on the kinases activated in these pathways, the IL-33-induced signalling is essential for production of IL-6 or IL-2. This was shown to control the dichotomy between RORγt+ and Helios+ Tregs , respectively. SCF, the ligand of c-Kit (CD117), can enhance these effects. Here, we show that IL-3, another growth factor for mast cells, is essential for the expression of ICOS-L on BMMCs, and costimulation with IL-3 potentiated the IL-33-induced IL-6 production similar to SCF. In contrast to the enhanced IL-2 production by SCF-induced modulation of the IL-33 signalling, IL-3 blocked the production of IL-2. Consequently, IL-3 shifted the IL-33-induced Treg dichotomy towards RORγt+ Tregs at the expense of RORγt- Helios+ Tregs . However, ICOS-L expression was downregulated by IL-33. In line with that, ICOS-L did not play any important role in the Treg modulation by IL-3/IL-33-activated mast cells. These findings demonstrate that different from the mast cell growth factor SCF, IL-3 can alter the IL-33-induced and mast cell-dependent regulation of Treg subpopulations by modulating mast cell-derived cytokine profiles.


Asunto(s)
Ligando Coestimulador de Linfocitos T Inducibles/metabolismo , Interleucina-33/farmacología , Interleucina-3/farmacología , Interleucina-6/metabolismo , Mastocitos/efectos de los fármacos , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Comunicación Paracrina/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Animales , Células Cultivadas , Técnicas de Cocultivo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mastocitos/inmunología , Mastocitos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Fenotipo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
5.
Allergy ; 76(4): 1109-1122, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32895937

RESUMEN

BACKGROUND: Vitamin A regulates the adaptive immune response and a modulatory impact on type I allergy is discussed. The cellular mechanisms are largely unknown. OBJECTIVE: To determine the vitamin A-responding specific lymphocyte reaction in vivo. METHODS: Antigen-specific B and T lymphocytes were analyzed in an adoptive transfer airway inflammation mouse model in response to 9-cis retinoic acid (9cRA) and after lymphocyte-specific genetic targeting of the receptor RARα. Flow cytometry, quantitative PCR, next-generation sequencing, and specific Ig-ELISA were used to characterize the cells functionally. RESULTS: Systemic 9cRA profoundly enhanced the specific IgA-secreting B-cell frequencies in the lung tissue and serum IgA while reducing serum IgE concentrations. RARα overexpression in antigen-specific B cells promoted differentiation into plasmablasts at the expense of germinal center B cells. In antigen-specific T cells, RARα strongly promoted the differentiation of T follicular helper cells followed by an enhanced germinal center response. CONCLUSIONS: 9cRA signaling via RARα impacts the allergen-specific immunoglobulin response directly by the differentiation of B cells and indirectly by promoting T follicular helper cells.


Asunto(s)
Hipersensibilidad , Vitamina A , Animales , Diferenciación Celular , Centro Germinal , Ratones , Células T Auxiliares Foliculares , Linfocitos T Colaboradores-Inductores
6.
J Immunol ; 202(8): 2229-2239, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30833348

RESUMEN

T follicular helper (Tfh) cells are a specialized T cell subset that regulates the long-lived production of highly specific Abs by B cells during the germinal center (GC) reaction. However, the transcriptional network sustaining the Tfh cell phenotype and function is still incompletely understood. In this study, we identify the transcription factor Bach2 as a central negative regulator of Tfh cells. Ectopic overexpression of Bach2 in murine Tfh cells resulted in a rapid loss of their phenotype and subsequent breakdown of the GC response. Low Bach2 expression levels are required to maintain high expression of the signature cytokine IL-21, the coinhibitory receptor TIGIT and the transcriptional repressor Bcl-6. In stark contrast to the regulatory network in GC B cells, Bach2 in Tfh cells is not coexpressed with Bcl-6 at high levels to inhibit the antagonizing factor Blimp-1, but suppresses Bcl-6 by direct binding to the promoter. These data reveal that by replacing an activating complex of Batf and Irf-4 at the Bcl-6 promoter, Bach2 regulates the transcriptional network of Tfh cells in a different way, as in GC B cells.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Regulación de la Expresión Génica/inmunología , Centro Germinal/inmunología , Proteínas Proto-Oncogénicas c-bcl-6 , Linfocitos T Reguladores/inmunología , Transcripción Genética/inmunología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/inmunología , Centro Germinal/citología , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/inmunología , Interleucinas/genética , Interleucinas/inmunología , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas/inmunología , Proteínas Proto-Oncogénicas c-bcl-6/genética , Proteínas Proto-Oncogénicas c-bcl-6/inmunología , Linfocitos T Reguladores/citología
7.
J Immunol ; 199(7): 2305-2315, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28814605

RESUMEN

The proper regulation of ICOS and ICOS ligand (ICOSL) has been shown to be essential for maintaining proper immune homeostasis. Loss of either protein results in defective humoral immunity, and overexpression of ICOS results in aberrant Ab production resembling lupus. How ICOSL is regulated in response to ICOS interaction is still unclear. We demonstrate that a disintegrin and metalloproteinase (ADAM)10 is the primary physiological sheddase of ICOSL in mice and humans. Using an in vivo system in which ADAM10 is deleted only on B cells, elevated levels of ICOSL were seen. This increase is also seen when ADAM10 is deleted from human B cell lines. Identification of the primary sheddase has allowed the characterization of a novel mechanism of ICOS regulation. In wild-type mice, interaction of ICOS/ICOSL results in ADAM10-induced shedding of ICOSL on B cells and moderate ICOS internalization on T cells. When this shedding is blocked, excessive ICOS internalization occurs. This results in severe defects in T follicular helper development and TH2 polarization, as seen in a house dust mite exposure model. In addition, enhanced TH1 and TH17 immune responses are seen in experimental autoimmune encephalomyelitis. Blockade of ICOSL rescues T cell ICOS surface expression and rescues, at least in part, T follicular helper numbers and the abnormal Ab production previously reported in these mice. Overall, we propose a novel regulation of the ICOS/ICOSL axis, with ADAM10 playing a direct role in regulating ICOSL, as well as indirectly regulating ICOS, thus controlling ICOS/ICOSL-dependent responses.


Asunto(s)
Linfocitos B/inmunología , Regulación de la Expresión Génica , Ligando Coestimulador de Linfocitos T Inducibles/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Proteína ADAM10/deficiencia , Proteína ADAM10/genética , Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/deficiencia , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Homeostasis , Humanos , Ligando Coestimulador de Linfocitos T Inducibles/genética , Ligando Coestimulador de Linfocitos T Inducibles/inmunología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Pyroglyphidae/inmunología , Células TH1/inmunología , Células Th17/inmunología
8.
Am J Respir Cell Mol Biol ; 59(4): 437-447, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29676593

RESUMEN

B-cell interaction with follicular helper T cells and subsequent differentiation of B cells into high-affinity APCs normally takes place in secondary lymphoid organs. The costimulator ICOS plays a key role in this process and is therefore considered as an attractive target to modulate exaggerated B-cell responses in autoimmune or allergic diseases. Inflamed tissues were recently recognized as additional sites of active T-cell/B-cell interaction. To analyze whether ICOS costimulation is also important there, we employed a mouse airway inflammation model that allows direct comparison of immune reactions in the lung-draining lymph node and the lung tissue as well as assessment of the relative importance of dendritic cells versus B cells as APCs. In both organs, ICOS regulated the pool size of antigen-specific T and B cells and B-cell differentiation into germinal center(-like) cells but not into antibody-secreting cells. In the lymph node, lack of ICOS costimulation drastically reduced the frequency of T follicular helper cells but did not affect production of T-helper cell type 2 (Th2) cytokines. Vice versa in the lung tissue, ICOS did not change PD-1 expression on infiltrating T cells but regulated Th2 cytokine production, a process for which ICOS ligand expression on B cells was of particular importance. Taken together, the results of this study show that ICOS differentially regulates effector T cells in secondary lymphoid organs and inflamed tissues but that blockade of the ICOS pathway is suitable to target T cell-dependent B cell responses at both sites.


Asunto(s)
Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Inflamación/patología , Tejido Linfoide/metabolismo , Linfocitos T/metabolismo , Animales , Células Presentadoras de Antígenos/metabolismo , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Eosinófilos/metabolismo , Inmunoglobulina A/metabolismo , Cambio de Clase de Inmunoglobulina , Inmunoglobulina E/metabolismo , Ligando Coestimulador de Linfocitos T Inducibles/metabolismo , Ganglios Linfáticos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía/patología , Receptor de Muerte Celular Programada 1/metabolismo
9.
PLoS Pathog ; 12(11): e1005999, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27812214

RESUMEN

Parasite-specific antibodies protect against blood-stage Plasmodium infection. However, in malaria-endemic regions, it takes many months for naturally-exposed individuals to develop robust humoral immunity. Explanations for this have focused on antigenic variation by Plasmodium, but have considered less whether host production of parasite-specific antibody is sub-optimal. In particular, it is unclear whether host immune factors might limit antibody responses. Here, we explored the effect of Type I Interferon signalling via IFNAR1 on CD4+ T-cell and B-cell responses in two non-lethal murine models of malaria, P. chabaudi chabaudi AS (PcAS) and P. yoelii 17XNL (Py17XNL) infection. Firstly, we demonstrated that CD4+ T-cells and ICOS-signalling were crucial for generating germinal centre (GC) B-cells, plasmablasts and parasite-specific antibodies, and likewise that T follicular helper (Tfh) cell responses relied on B cells. Next, we found that IFNAR1-signalling impeded the resolution of non-lethal blood-stage infection, which was associated with impaired production of parasite-specific IgM and several IgG sub-classes. Consistent with this, GC B-cell formation, Ig-class switching, plasmablast and Tfh differentiation were all impaired by IFNAR1-signalling. IFNAR1-signalling proceeded via conventional dendritic cells, and acted early by limiting activation, proliferation and ICOS expression by CD4+ T-cells, by restricting the localization of activated CD4+ T-cells adjacent to and within B-cell areas of the spleen, and by simultaneously suppressing Th1 and Tfh responses. Finally, IFNAR1-deficiency accelerated humoral immune responses and parasite control by boosting ICOS-signalling. Thus, we provide evidence of a host innate cytokine response that impedes the onset of humoral immunity during experimental malaria.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Inmunidad Humoral/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Malaria/inmunología , Receptor de Interferón alfa y beta/inmunología , Animales , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Plasmodium chabaudi/inmunología , Plasmodium yoelii/inmunología , Transducción de Señal/inmunología
10.
Immunity ; 30(2): 228-41, 2009 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-19217324

RESUMEN

During evolutionary adaptation in the immune system, host defense is traded off against autoreactivity. Signals through the costimulatory receptor CD28 enable T cells to respond specifically to pathogens, whereas those through the related costimulatory receptor, ICOS, which arose by gene duplication, are critical for affinity maturation and memory antibody responses. ICOS ligand, unlike the pathogen-inducible CD28 ligands, is widely and constitutively expressed in the immune system. Here, we show that crosstalk between these two pathways provides a mechanism for obviating the normal T cell dependence on CD28. Several CD28-mediated responses-generation of follicular helper T cells, germinal center formation, T helper 1 cell-dependent extrafollicular antibody responses to Salmonella and bacterial clearance, and regulatory T cell homeostasis-became independent of CD28 and dependent on ICOS when the E3 ubiquitin ligase Roquin was mutated. Mechanisms to functionally compartmentalize ICOS and CD28 signals are thus critical for two-signal control of normal immune reactions.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/inmunología , Antígenos CD28/inmunología , Linfocitos T/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Alelos , Animales , Antígenos de Diferenciación de Linfocitos T/genética , Antígenos de Diferenciación de Linfocitos T/metabolismo , Antígenos CD28/genética , Antígenos CD28/metabolismo , Diferenciación Celular/inmunología , Regulación de la Expresión Génica , Centro Germinal/inmunología , Memoria Inmunológica/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles , Interleucina-2/inmunología , Interleucina-2/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Salmonella/inmunología , Transducción de Señal/inmunología , Linfocitos T/citología , Linfocitos T/metabolismo , Timo/citología , Timo/inmunología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
11.
Immunity ; 31(5): 823-33, 2009 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-19913446

RESUMEN

The expression of the chemokine receptor XCR1 and the function of its ligand XCL1 (otherwise referred to as ATAC, lymphotactin, or SCM-1) remained elusive to date. In the present report we demonstrated that XCR1 is exclusively expressed on murine CD8(+) dendritic cells (DCs) and showed that XCL1 is a potent and highly specific chemoattractant for this DC subset. CD8(+) T cells abundantly secreted XCL1 8-36 hr after antigen recognition on CD8(+) DCs in vivo, in a period in which stable T cell-DC interactions are known to occur. Functionally, XCL1 increased the pool of antigen-specific CD8(+) T cells and their capacity to secrete IFN-gamma. Absence of XCL1 impaired the development of cytotoxicity to antigens cross-presented by CD8(+) DCs. The XCL1-XCR1 axis thus emerges as an integral component in the development of efficient cytotoxic immunity in vivo.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Regulación de la Expresión Génica/inmunología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Linfocitos B/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Quimiocinas C/farmacología , Células Dendríticas/efectos de los fármacos , Humanos , Inmunohistoquímica , Células Asesinas Naturales/inmunología , Ratones , Ratones Noqueados , Receptores Acoplados a Proteínas G/genética , Bazo/citología
12.
J Immunol ; 194(3): 1069-79, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25520399

RESUMEN

Current subunit vaccines are incapable of inducing Ag-specific CD8(+) T cell cytotoxicity needed for the defense of certain infections and for therapy of neoplastic diseases. In experimental vaccines, cytotoxic responses can be elicited by targeting of Ag into cross-presenting dendritic cells (DC), but almost all available systems use target molecules also expressed on other cells and thus lack the desired specificity. In the present work, we induced CD8(+) T cell cytotoxicity by targeting of Ag to XCR1, a chemokine receptor exclusively expressed on murine and human cross-presenting DC. Targeting of Ag with a mAb or the chemokine ligand XCL1 was highly specific, as determined with XCR1-deficient mice. When applied together with an adjuvant, both vector systems induced a potent cytotoxic response preventing the outgrowth of an inoculated aggressive tumor. By generating a transgenic mouse only expressing the human XCR1 on its cross-presenting DC, we could demonstrate that targeting of Ag using human XCL1 as vector is fully effective in vivo. The specificity and efficiency of XCR1-mediated Ag targeting to cross-presenting DC, combined with its lack of adverse effects, make this system a prime candidate for the development of therapeutic cytotoxic vaccines in humans.


Asunto(s)
Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Receptores de Quimiocina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Vacunas contra el Cáncer/inmunología , Diferenciación Celular , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Humanos , Cambio de Clase de Inmunoglobulina , Activación de Linfocitos/inmunología , Ratones , Ratones Transgénicos , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Unión Proteica , Receptores de Quimiocina/genética , Receptores Acoplados a Proteínas G/genética , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Carga Tumoral
13.
Eur J Immunol ; 45(10): 2766-72, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26249010

RESUMEN

Group 2 innate lymphoid cells (ILC2s) are innate effectors playing an important role in the defense against helminthic infections and in the pathogenesis of allergic inflammation. Cytokines have been identified as the major stimuli driving ILC2 activation and expansion. Conversely, it is unclear whether costimulatory molecules contribute to regulation of ILC2 functions. ILC2s display high expression of inducible T-cell costimulator (ICOS), which belongs to the CD28 superfamily, and which has been shown to control late effector T-cell functions, and is of utmost importance for the humoral immune response. However, the biological function of ICOS expression on ILC2s is unknown. Here, we show that ICOS signaling in mice regulates ILC2 homeostasis independently of T cells and B cells, by promoting proliferation and accumulation of mature ILC2s in lung and intestine. In a model of IL-33-induced airway inflammation, ICOS controls ILC2 activation and eosinophil infiltration in the lung. Our data identify a role of ICOS in innate immunity and indicate that not only cytokines, but also costimulatory pathways such as those involving ICOS, can contribute to regulate the ILC2 pool. Thus, ICOS costimulation blockade, which is currently under clinical evaluation for inhibiting the humoral immune response, could also target innate inflammatory circuits.


Asunto(s)
Homeostasis/inmunología , Inmunidad Humoral/fisiología , Inmunidad Innata/fisiología , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Intestinos/inmunología , Pulmón/inmunología , Animales , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Inflamación/genética , Inflamación/inmunología , Interleucina-33/genética , Interleucina-33/inmunología , Intestinos/citología , Pulmón/citología , Ratones , Ratones Mutantes , Linfocitos T/citología , Linfocitos T/inmunología
14.
J Immunol ; 193(6): 2753-63, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25108021

RESUMEN

Immune cells regulate cell surface receptor expression during their maturation, activation, and motility. Although many of these receptors are regulated largely at the level of expression, protease-mediated ectodomain shedding represents an alternative means of refashioning the surface of immune cells. Shedding is largely attributed to a family of a disintegrin and metalloprotease domain (ADAM) metalloproteases, including ADAM17. Although ADAM17 is well known to contribute to the innate immune response, mainly by releasing TNF-α, much less is known about whether/how this metalloprotease regulates adaptive immunity. To determine whether ADAM17 contributes to regulating adaptive immune responses, we took advantage of ADAM17 hypomorphic (ADAM17(ex/ex)) mice, in which ADAM17 expression is reduced by 90-95% compared with wild-type littermates. In this study, we show that that ADAM17 deficiency results in spleen and lymph node enlargement, as well as increased levels of Ag-specific class-switched Ig production following immunization with OVA together with anti-CD40 mAbs and polyinosinic-polycytidylic acid. Moreover, we demonstrate that the costimulatory ligand ICOS ligand (ICOSL) is selectively downregulated on the surface of B cells in an ADAM17-specific manner, although it is not proteolitically processed by recombinant ADAM17 in vitro. Finally, we show that higher cell surface levels of ICOSL in ADAM17(ex/ex) mice may contribute to the development of excessive Ab responses. Therefore, our data suggest a functional link between ADAM17 and ICOSL in controlling adaptive immune responses.


Asunto(s)
Proteínas ADAM/inmunología , Linfocitos B/inmunología , Inmunidad Humoral , Ligando Coestimulador de Linfocitos T Inducibles/inmunología , Proteínas ADAM/genética , Proteína ADAM17 , Animales , Anticuerpos Monoclonales/inmunología , Formación de Anticuerpos/inmunología , Linfocitos B/trasplante , Antígenos CD40/inmunología , Células Cultivadas , Femenino , Cambio de Clase de Inmunoglobulina , Isotipos de Inmunoglobulinas/biosíntesis , Isotipos de Inmunoglobulinas/inmunología , Ligando Coestimulador de Linfocitos T Inducibles/biosíntesis , Ganglios Linfáticos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina/inmunología , Poli I-C/inmunología , Bazo/patología
15.
J Immunol ; 189(1): 234-44, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22661090

RESUMEN

We have established a comprehensive in vivo mouse model for the CD4(+) T cell response to an "innocuous" versus "dangerous" exogenous Ag and developed an in vivo test for tolerance. In this model, specific gene-expression signatures, distinctive upregulation of early T cell-communication molecules, and differential expansion of effector T cells (Teff) and regulatory T cells (Treg) were identified as central correlates of T cell tolerance and T cell immunity. Different from essentially all other T cell-activation molecules, ICOS was found to be induced in the immunity response and not by T cells activated under tolerogenic conditions. If expressed, ICOS did not act as a general T cell costimulator but selectively caused a massive expansion of effector CD4(+) T cells, leaving the regulatory CD4(+) T cell compartment largely undisturbed. Thus, ICOS strongly contributed to the dramatic change in the balance between Ag-specific Teff and Treg from ∼1:1 at steady state to 21:1 at the height of the immune response. This newly defined role for the balance of Teff to Treg, together with its known key function in T cell help for B cells, establishes ICOS as a central mediator of immunity. Given its exceptionally selective induction on CD4(+) T cells under inflammatory, but not tolerogenic, conditions, ICOS emerges as a pivotal effector molecule in the early decision between tolerance and immunity to exogenous Ag.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Tolerancia Inmunológica , Proteína Coestimuladora de Linfocitos T Inducibles/fisiología , Inmunidad Adaptativa/genética , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Tolerancia Inmunológica/genética , Proteína Coestimuladora de Linfocitos T Inducibles/deficiencia , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
16.
Nat Commun ; 15(1): 4182, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38755157

RESUMEN

Bone marrow plasma cells (BMPC) are the correlate of humoral immunity, consistently releasing antibodies into the bloodstream. It remains unclear if BMPC reflect different activation environments or maturation of their precursors. Here we define human BMPC heterogeneity and track the recruitment of antibody-secreting cells (ASC) from SARS-CoV-2 vaccine immune reactions to the bone marrow (BM). Trajectories based on single-cell transcriptomes and repertoires of peripheral and BM ASC reveal sequential colonisation of BMPC compartments. In activated B cells, IL-21 suppresses CD19 expression, indicating that CD19low-BMPC are derived from follicular, while CD19high-BMPC originate from extrafollicular immune reactions. In primary immune reactions, both CD19low- and CD19high-BMPC compartments are populated. In secondary immune reactions, most BMPC are recruited to CD19high-BMPC compartments, reflecting their origin from extrafollicular reactivations of memory B cells. A pattern also observable in vaccinated-convalescent individuals and upon diphtheria/tetanus/pertussis recall-vaccination. Thus, BMPC diversity reflects the evolution of a given humoral immune response.


Asunto(s)
Antígenos CD19 , Médula Ósea , Interleucinas , Células Plasmáticas , Humanos , Células Plasmáticas/inmunología , Interleucinas/inmunología , Interleucinas/metabolismo , Médula Ósea/inmunología , Antígenos CD19/inmunología , Antígenos CD19/metabolismo , Inmunidad Humoral/inmunología , COVID-19/inmunología , COVID-19/virología , SARS-CoV-2/inmunología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/citología , Análisis de la Célula Individual , Adulto , Linfocitos B/inmunología , Células Productoras de Anticuerpos/inmunología , Femenino , Masculino , Vacunación , Persona de Mediana Edad , Vacuna contra Difteria, Tétanos y Tos Ferina/inmunología
17.
Eur J Immunol ; 42(8): 1981-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22730020

RESUMEN

T-follicular helper (TFH) cells represent the subpopulation of CD4(+) T cells that provides help for antigen-specific B cells in the GC response. They are generated from naïve T cells during an immune response and are imprinted by their master transcription factor Bcl-6. It has been a long-standing question if TFH cells contribute to the CD4(+) memory pool after the GC response has been terminated. To answer this question, we sorted antigen-specific TFH and non-TFH effector cells from an ongoing GC response and transferred them into naïve mice. Without further signals via the TCR, transferred cells rapidly contracted with a small population of both TFH and non-TFH cells surviving as memory cells in peripheral lymphoid organs for at least 4 weeks in the absence of antigen. TFH cells strongly downregulated their signature genes Bcl-6, CXCR5, and PD-1 in the memory phase. Upon rechallenge with antigen they rapidly upregulated these markers again. An enhanced potential to produce IL-21, paired with higher expression of CXCR5 and lower expression of CCR7, should enable TFH memory cells to provide more efficient help for antigen-specific B cells than their non-TFH counterparts.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Memoria Inmunológica , Proteínas Proto-Oncogénicas c-bcl-6/biosíntesis , Receptores CXCR5/biosíntesis , Linfocitos T Colaboradores-Inductores/inmunología , Traslado Adoptivo , Animales , Antígenos de Diferenciación/inmunología , Diferenciación Celular , Regulación hacia Abajo , Centro Germinal/inmunología , Interleucinas/biosíntesis , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor de Muerte Celular Programada 1 , Proteínas Proto-Oncogénicas c-bcl-6/inmunología , Receptores CCR7/biosíntesis , Receptores CXCR5/inmunología , Bazo/inmunología , Regulación hacia Arriba
18.
Nature ; 450(7167): 299-303, 2007 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-18172933

RESUMEN

Immune responses are normally targeted against microbial pathogens and not self-antigens by mechanisms that are only partly understood. Here we define a newly discovered pathway that prevents autoimmunity by limiting the levels on T lymphocytes of aco-stimulatory receptor, the inducible T-cell co-stimulator(ICOS). In sanroque mice homozygous for an M199R mutation in the ROQ domain of Roquin (also known as Rc3h1), increased Icos expression on T cells causes the accumulation of lymphocytes that is associated with a lupus-like autoimmune syndrome. Roquin normally limits Icos expression by promoting the degradation of Icos messenger RNA.A conserved segment in the unusually long ICOS 3' untranslated mRNA is essential for regulation by Roquin. This segment comprises a 47-base-pair minimal region complementary to T-cell-expressed microRNAs including miR-101, the repressive activity of which is disrupted by base-pair inversions predicted to abrogate miR-101 binding. These findings illuminate a critical post-transcriptional pathway within T cells that regulates lymphocyte accumulation and autoimmunity, and highlights the therapeutic potential of partially antagonising the ICOS pathway.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/genética , Autoinmunidad/genética , Autoinmunidad/inmunología , Regulación de la Expresión Génica , ARN Mensajero/biosíntesis , Linfocitos T/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Regiones no Traducidas 3'/genética , Regiones no Traducidas 3'/metabolismo , Animales , Antígenos de Diferenciación de Linfocitos T/metabolismo , Secuencia de Bases , Línea Celular , Humanos , Proteína Coestimuladora de Linfocitos T Inducibles , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Mutación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Secuencias Reguladoras de Ácido Ribonucleico/genética , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética
19.
J Allergy Clin Immunol ; 129(6): 1647-55.e13, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22502800

RESUMEN

BACKGROUND: Under inflammatory conditions, T cell-dependent (TD) protein antigens induce proinflammatory T- and B-cell responses. In contrast, tolerance induction by TD antigens without costimulation triggers the development of regulatory T cells. Under both conditions, IgG antibodies are generated, but whether they have different immunoregulatory functions remains elusive. OBJECTIVE: It was shown recently that proinflammatory or anti-inflammatory effector functions of IgG molecules are determined by different Fc N-linked glycosylation patterns. We sought to examine the Fc glycosylation and anti-inflammatory quality of IgG molecules formed on TD tolerance induction. METHODS: We administered chicken ovalbumin (OVA) with or without costimulus to mice and analyzed OVA-reactive IgG Fc glycosylation. The anti-inflammatory function of differentially glycosylated anti-OVA IgGs was further investigated in studies with dendritic cell cultures and in an in vivo model of allergic airway disease. Additionally, we analyzed the Fc glycosylation pattern of birch pollen-reactive serum IgGs after successful allergen-specific immunotherapy in patients. RESULTS: Stimulation with TD antigens under inflammatory conditions induces plasma cells expressing low levels of α2,6-sialyltransferase and producing desialylated IgGs. In contrast, plasma cells induced on tolerance induction did not downregulate α2,6-sialyltransferase expression and secreted immunosuppressive sialylated IgGs that were sufficient to block antigen-specific T- and B-cell responses, dendritic cell maturation, and allergic airway inflammation. Importantly, successful specific immunotherapy in allergic patients also induced sialylated allergen-specific IgGs. CONCLUSIONS: Our data show a novel antigen-specific immunoregulatory mechanism mediated by anti-inflammatory sialylated IgGs that are formed on TD tolerance induction. These findings might help to develop novel antigen-specific therapies for the treatment of allergy and autoimmunity.


Asunto(s)
Antígenos/inmunología , Tolerancia Inmunológica/inmunología , Inmunoglobulina G/inmunología , Linfocitos T/inmunología , Animales , Complejo Antígeno-Anticuerpo/inmunología , Desensibilización Inmunológica , Epítopos/inmunología , Femenino , Humanos , Hipersensibilidad/inmunología , Hipersensibilidad/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Receptores de IgG/metabolismo , Sialiltransferasas/biosíntesis , beta-D-Galactósido alfa 2-6-Sialiltransferasa
20.
Front Cell Dev Biol ; 11: 1256127, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38020883

RESUMEN

Introduction: Removal of poorly perfused capillaries by pruning contributes to remodeling the microvasculature to optimize oxygen and nutrient delivery. Blood flow drives this process by promoting the intravascular migration of endothelial cells in developing networks, such as in the yolk sac, zebrafish brain or postnatal mouse retina. Methods: In this study, we have implemented innovative tools to recognize capillary pruning in the complex 3D coronary microvasculature of the postnatal mouse heart. We have also experimentally tested the impact of decreasing pruning on the structure and function of this network by altering blood flow with two different vasodilators: losartan and prazosin. Results: Although both drugs reduced capillary pruning, a combination of experiments based on ex vivo imaging, proteomics, electron microscopy and in vivo functional approaches showed that losartan treatment resulted in an inefficient coronary network, reduced myocardial oxygenation and metabolic changes that delayed the arrest of cardiomyocyte proliferation, in contrast to the effects of prazosin, probably due to its concomitant promotion of capillary expansion. Discussion: Our work demonstrates that capillary pruning contributes to proper maturation and function of the heart and that manipulation of blood flow may be a novel strategy to refine the microvasculature and improve tissue perfusion after damage.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA