Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Environ Microbiol ; 24(12): 5924-5935, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35799468

RESUMEN

Endosymbiosis, an interaction between two species where one lives within the other, has evolved multiple times independently, but the underlying mechanisms remain unclear. Evolutionary theory suggests that for an endosymbiotic interaction to remain stable over time, births of both partners should be higher than their deaths in symbiosis and deaths of both partners should be higher than their births when living independently. However, experimentally measuring this can be difficult and conclusions tend to focus on the host. Using a ciliate-algal system (Paramecium bursaria host and Chlorella endosymbionts), we estimated the benefits and costs of endosymbiosis for both organisms using fitness measurements in different biotic environments to test under which environmental conditions the net effects of the interaction were positive for both partners. We found that the net effects of harbouring endosymbionts were positive for the ciliate hosts as it allowed them to survive in conditions of low-quality bacteria food. The algae benefitted by being endosymbiotic when predators such as the hosts were present, but the net effects were dependent on the total density of hosts, decreasing as hosts densities increased. Overall, we show that including context-dependency of endosymbiosis is essential in understanding how these interactions have evolved.


Asunto(s)
Chlorella , Cilióforos , Paramecium , Simbiosis , Análisis Costo-Beneficio
2.
Proc Natl Acad Sci U S A ; 108(24): 9927-32, 2011 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-21628593

RESUMEN

Cross-presentation of IgG-containing immune complexes (ICs) is an important means by which dendritic cells (DCs) activate CD8(+) T cells, yet it proceeds by an incompletely understood mechanism. We show that monocyte-derived CD8(-)CD11b(+) DCs require the neonatal Fc receptor for IgG (FcRn) to conduct cross-presentation of IgG ICs. Consequently, in the absence of FcRn, Fcγ receptor (FcγR)-mediated antigen uptake fails to initiate cross-presentation. FcRn is shown to regulate the intracellular sorting of IgG ICs to the proper destination for such cross-presentation to occur. We demonstrate that FcRn traps antigen and protects it from degradation within an acidic loading compartment in association with the rapid recruitment of key components of the phagosome-to-cytosol cross-presentation machinery. This unique mechanism thus enables cross-presentation to evolve from an atypically acidic loading compartment. FcRn-driven cross-presentation is further shown to control cross-priming of CD8(+) T-cell responses in vivo such that during chronic inflammation, FcRn deficiency results in inadequate induction of CD8(+) T cells. These studies thus demonstrate that cross-presentation in CD8(-)CD11b(+) DCs requires a two-step mechanism that involves FcγR-mediated internalization and FcRn-directed intracellular sorting of IgG ICs. Given the centrality of FcRn in controlling cross-presentation, these studies lay the foundation for a unique means to therapeutically manipulate CD8(+) T-cell responses.


Asunto(s)
Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoglobulina G/inmunología , Receptores Fc/inmunología , Animales , Antígenos/inmunología , Western Blotting , Antígeno CD11b/inmunología , Antígeno CD11b/metabolismo , Antígenos CD8/inmunología , Antígenos CD8/metabolismo , Colitis/inducido químicamente , Colitis/inmunología , Colitis/metabolismo , Citosol/inmunología , Citosol/metabolismo , Células Dendríticas/metabolismo , Sulfato de Dextran , Citometría de Flujo , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Concentración de Iones de Hidrógeno , Inmunoglobulina G/genética , Inmunoglobulina G/metabolismo , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mutación , NADPH Oxidasa 2 , NADPH Oxidasas/inmunología , NADPH Oxidasas/metabolismo , Fagosomas/inmunología , Fagosomas/metabolismo , Unión Proteica , Receptores Fc/genética , Receptores Fc/metabolismo , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , ATPasas de Translocación de Protón Vacuolares/inmunología , ATPasas de Translocación de Protón Vacuolares/metabolismo , Proteínas de Unión al GTP rab/inmunología , Proteínas de Unión al GTP rab/metabolismo , Proteínas rab27 de Unión a GTP
3.
J Immunol ; 187(1): 164-71, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21622859

RESUMEN

The IgE-mediated and Th2-dependent late-phase reaction remains a mechanistically enigmatic and daunting element of human allergic inflammation. In this study, we uncover the FcεRI on dendritic cells (DCs) as a key in vivo component of this form of allergy. Because rodent, unlike human, DCs lack FcεRI, this mechanism could be revealed only by using a new transgenic mouse model with human-like FcεRI expression on DCs. In the presence of IgE and allergen, FcεRI(+) DCs instructed naive T cells to differentiate into Th2 cells in vitro and boosted allergen-specific Th2 responses and Th2-dependent eosinophilia at the site of allergen exposure in vivo. Thus, FcεRI on DCs drives the cascade of pathogenic reactions linking the initial allergen capture by IgE with subsequent Th2-dominated T cell responses and the development of late-phase allergic tissue inflammation.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/patología , Mediadores de Inflamación/metabolismo , Receptores de IgE/metabolismo , Células Th2/inmunología , Células Th2/patología , Alérgenos/toxicidad , Animales , Antígenos de Plantas/toxicidad , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/metabolismo , Femenino , Humanos , Mediadores de Inflamación/fisiología , Mediadores de Inflamación/toxicidad , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/toxicidad , Unión Proteica/genética , Unión Proteica/inmunología , Receptores de IgE/deficiencia , Receptores de IgE/fisiología , Células Th2/metabolismo , Factores de Tiempo
4.
Cancer Immunol Immunother ; 61(9): 1521-5, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22042251

RESUMEN

One of the goals of cell-based immune therapy in cancer is the induction of tumor-specific cytotoxic T-lymphocyte (CTL) responses. To achieve this objective, the ability of dendritic cells (DC) to cross-present tumor antigens can be exploited. One of the most efficient pathways for the induction of CTLs by cross-presentation is mediated by immunoglobulins of the IgG class, which are used by DCs to sample antigen in the form of immune complexes via Fc-gamma receptors. Could DCs use an IgE-mediated cross-presentation mechanism in a comparable manner to induce CTLs? We here discuss the potential of two human IgE Fc receptors, FcεRI and FcεRII, to serve as antigen uptake receptors for IgE-mediated cross-presentation. We conclude that the existence of an IgE-mediated cross-presentation pathway would provide a direct link between IgE-driven immune responses and CTL activity.


Asunto(s)
Células Dendríticas/inmunología , Inmunoglobulina E/inmunología , Receptores de IgG/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Presentación de Antígeno/inmunología , Reactividad Cruzada/inmunología , Células Dendríticas/metabolismo , Humanos , Inmunoglobulina E/metabolismo , Receptores de IgG/metabolismo , Linfocitos T Citotóxicos/metabolismo
5.
J Immunol ; 185(6): 3217-26, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20733204

RESUMEN

The major cellular antioxidant glutathione is depleted during HIV infection and in obesity. Although the consequence of glutathione depletion on immune function is starting to emerge, it is currently not known whether glutathione dysregulation influences the differentiation and maturation of dendritic cells (DCs). Moreover, the effect of glutathione depletion on DC effector functions, such as Ag presentation, is poorly understood. Glutathione synthesis depends on the cystine/glutamate antiporter, which transports the rate-limiting precursor cystine into the cell in exchange for glutamate. In this paper, we present a detailed study of antiporter function in DCs and demonstrate a role for the antiporter in DC differentiation and cross-presentation. We show that the antiporter is the major mechanism for transport of cystine and glutamate and modulates the intracellular glutathione content and glutathione efflux from DCs. Blocking antiporter-dependent cystine transport decreases intracellular glutathione levels, and these effects correlate with reduced transcription of the functional subunit of the antiporter. We further demonstrate that blocking antiporter activity interferes with DC differentiation from monocyte precursors, but antiporter activity is not required for LPS-induced phenotypic maturation. Finally, we show that inhibiting antiporter uptake of cystine interferes with presentation of exogenous Ag to class II MHC-restricted T cells and blocks cross-presentation on MHC class I. We conclude that aberrant antiporter function disrupts glutathione homeostasis in DCs and may contribute to impaired immunity in the diseased host.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/fisiología , Presentación de Antígeno/inmunología , Diferenciación Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Sistema de Transporte de Aminoácidos y+/antagonistas & inhibidores , Animales , Presentación de Antígeno/genética , Transporte Biológico/inmunología , Diferenciación Celular/genética , Células Cultivadas , Reactividad Cruzada/genética , Reactividad Cruzada/inmunología , Cistina/metabolismo , Células Dendríticas/metabolismo , Ácido Glutámico/metabolismo , Glutatión/antagonistas & inhibidores , Glutatión/metabolismo , Homeostasis/inmunología , Humanos , Líquido Intracelular/inmunología , Líquido Intracelular/metabolismo , Lipopolisacáridos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovalbúmina/inmunología , Ovalbúmina/metabolismo
6.
J Biol Chem ; 285(20): 15314-15323, 2010 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-20304923

RESUMEN

The high affinity receptor for IgE, Fc epsilon receptor I (FcepsilonRI), is an activating immune receptor and key regulator of allergy. Antigen-mediated cross-linking of IgE-loaded FcepsilonRI alpha-chains induces cell activation via immunoreceptor tyrosine-based activation motifs in associated signaling subunits, such as FcepsilonRI gamma-chains. Here we show that the human FcepsilonRI alpha-chain can efficiently reach the cell surface by itself as an IgE-binding receptor in the absence of associated signaling subunits when the endogenous signal peptide is swapped for that of murine major histocompatibility complex class-I H2-K(b). This single-chain isoform of FcepsilonRI exited the endoplasmic reticulum (ER), trafficked to the Golgi and, subsequently, trafficked to the cell surface. Mutational analysis showed that the signal peptide regulates surface expression in concert with other described ER retention signals of FcepsilonRI-alpha. Once the FcepsilonRI alpha-chain reached the cell surface by itself, it formed a ligand-binding receptor that stabilized upon IgE contact. Independently of the FcepsilonRI gamma-chain, this single-chain FcepsilonRI was internalized after receptor cross-linking and trafficked into a LAMP-1-positive lysosomal compartment like multimeric FcepsilonRI. These data suggest that the single-chain isoform is capable of shuttling IgE-antigen complexes into antigen loading compartments, which plays an important physiologic role in the initiation of immune responses toward allergens. We propose that, in addition to cytosolic and transmembrane ER retention signals, the FcepsilonRI alpha-chain signal peptide contains a negative regulatory signal that prevents expression of an immunoreceptor tyrosine-based activation motif-free IgE receptor pool, which would fail to induce cell activation.


Asunto(s)
Señales de Clasificación de Proteína , Receptores de IgE/metabolismo , Tirosina/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Secuencia de Bases , Línea Celular , Membrana Celular/metabolismo , Cartilla de ADN , Retículo Endoplásmico/metabolismo , Citometría de Flujo , Aparato de Golgi/metabolismo , Humanos , Microscopía Fluorescente , Datos de Secuencia Molecular , Transporte de Proteínas , Receptores de IgE/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
J Immunol ; 183(1): 66-74, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19535631

RESUMEN

The transcription factor aryl hydrocarbon receptor (AhR) represents a promising therapeutic target in allergy and autoimmunity. AhR signaling induced by the newly described ligand VAF347 inhibits allergic lung inflammation as well as suppresses pancreatic islet allograft rejection. These effects are likely mediated via alterations in dendritic cell (DC) function. Moreover, VAF347 induces tolerogenic DCs. Langerhans cells (LCs) are immediate targets of exogenous AhR ligands at epithelial surfaces; how they respond to AhR ligands remained undefined. We studied AhR expression and function in human LCs and myelopoietic cell subsets using a lineage differentiation and gene transduction model of human CD34(+) hematopoietic progenitors. We found that AhR is highly regulated during myeloid subset differentiation. LCs expressed highest AhR levels followed by monocytes. Conversely, neutrophil granulocytes lacked AhR expression. AhR ligands including VAF347 arrested the differentiation of monocytes and LCs at an early precursor cell stage, whereas progenitor cell expansion or granulopoiesis remained unimpaired. AhR expression was coregulated with the transcription factor PU.1 during myeloid subset differentiation. VAF347 inhibited PU.1 induction during initial monocytic differentiation, and ectopic PU.1 restored monocyte and LC generation in the presence of this compound. AhR ligands failed to interfere with cytokine receptor signaling during LC differentiation and failed to impair LC activation/maturation. VAF347-mediated antiproliferative effect on precursors undergoing LC lineage differentiation occurred in a clinically applicable serum-free culture model and was not accompanied by apoptosis induction. In conclusion, AhR agonist signaling interferes with transcriptional processes leading to monocyte/DC lineage commitment of human myeloid progenitor cells.


Asunto(s)
Diferenciación Celular/inmunología , Células de Langerhans/inmunología , Células de Langerhans/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Inhibidores de Crecimiento/farmacología , Inhibidores de Crecimiento/fisiología , Humanos , Células K562 , Células de Langerhans/citología , Monocitos/citología , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/inmunología , Células Progenitoras Mieloides/metabolismo , Pirimidinas/farmacología , Receptores de Hidrocarburo de Aril/agonistas , Receptores de Hidrocarburo de Aril/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Transcripción Genética/efectos de los fármacos , Transcripción Genética/inmunología
8.
Cancer Res ; 81(11): 3079-3091, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33504557

RESUMEN

p53 is a transcription factor that plays a central role in guarding the genomic stability of cells through cell-cycle arrest or induction of apoptosis. However, the effects of p53 in antitumor immunity are poorly understood. To investigate the role of p53 in controlling tumor-immune cell cross-talk, we studied murine syngeneic models treated with HDM201, a potent and selective second-generation MDM2 inhibitor. In response to HDM201 treatment, the percentage of dendritic cells increased, including the CD103+ antigen cross-presenting subset. Furthermore, HDM201 increased the percentage of Tbet+Eomes+ CD8+ T cells and the CD8+/Treg ratio within the tumor. These immunophenotypic changes were eliminated with the knockout of p53 in tumor cells. Enhanced expression of CD80 on tumor cells was observed in vitro and in vivo, which coincided with T-cell-mediated tumor cell killing. Combining HDM201 with PD-1 or PD-L1 blockade increased the number of complete tumor regressions. Responding mice developed durable, antigen-specific memory T cells and rejected subsequent tumor implantation. Importantly, antitumor activity of HDM201 in combination with PD-1/PD-L1 blockade was abrogated in p53-mutated and knockout syngeneic tumor models, indicating the effect of HDM201 on the tumor is required for triggering antitumor immunity. Taken together, these results demonstrate that MDM2 inhibition triggers adaptive immunity, which is further enhanced by blockade of PD-1/PD-L1 pathway, thereby providing a rationale for combining MDM2 inhibitors and checkpoint blocking antibodies in patients with wild-type p53 tumors. SIGNIFICANCE: This study provides a mechanistic rationale for combining checkpoint blockade immunotherapy with MDM2 inhibitors in patients with wild-type p53 tumors.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Neoplasias del Colon/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Células del Estroma/inmunología , Microambiente Tumoral/inmunología , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Animales , Apoptosis , Proliferación Celular , Neoplasias del Colon/inmunología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Quimioterapia Combinada , Femenino , Humanos , Imidazoles/farmacología , Inhibidores de Puntos de Control Inmunológico/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Desnudos , Pirimidinas/farmacología , Pirroles/farmacología , Células del Estroma/efectos de los fármacos , Células Tumorales Cultivadas , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Exp Hematol ; 32(3): 270-6, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15003312

RESUMEN

Myeloperoxidase (MPO) represents an early-appearing and highly reliable intracellular myeloid lineage marker molecule. MPO protein can be detected in a subset of human hematopoietic bone marrow progenitor cells and in granulomonopoietic (GM) cells. However, other myeloid-related cell types such as epidermal Langerhans-type dendritic cells (LC) lack MPO. Therefore, human myeloid progenitors might be subdivided based on MPO protein expression into functional subsets. Here we identified two consecutive myelopoietic cell stages, i.e., early myeloid progenitors that lack MPO, as well as their immediate MPO+ progeny. MPO- myeloid progenitors possess previously described granulomonocyte (GM) progenitor-associated cell-surface characteristics (CD34+CD45RA+CD13+lin-). They are specifically recruited and can be expanded in cultures of CD34+ cord blood cells in response to early-acting hematopoietic cytokines. Furthermore, cell fractions enriched in MPO- myeloid progenitors efficiently developed along Langerhans-type dendritic cell (LC) and granulomonocytic (GM) lineages, whereas progeny enriched in MPO+ cells showed diminished LC potential. In line with this, peripheral blood progenitors, known to possess LC differentiation potential, lacked MPO expression. We conclude that differential expression of MPO therefore further characterizes cells with myeloid or LC potential.


Asunto(s)
Células Dendríticas/enzimología , Hematopoyesis , Células Mieloides/enzimología , Peroxidasa/análisis , Antígenos CD34/análisis , Biomarcadores , Diferenciación Celular , Linaje de la Célula , Separación Celular , Células Cultivadas/citología , Células Cultivadas/enzimología , Células Dendríticas/citología , Inducción Enzimática , Sangre Fetal/citología , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunofenotipificación , Lactoferrina/análisis , Células de Langerhans/citología , Células de Langerhans/enzimología , Antígenos Comunes de Leucocito/análisis , Receptores de Lipopolisacáridos/análisis , Células Mieloides/clasificación , Células Mieloides/citología , Especificidad de Órganos , Peroxidasa/genética
10.
Mol Immunol ; 68(2 Pt A): 116-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26052071

RESUMEN

Immunoglobulin E (IgE) functions as an Fc-receptor-bound antigen sensor for mast cells and basophils, the classical effector cells of allergy. A cell-bound IgE pool is formed when monomeric IgE binds to FcɛRI, the high affinity IgE Fc receptor on these cells, and minor amounts of antigen are sufficient to trigger the pro-allergic innate IgE effector axis. Additionally, FcɛRI is constitutively expressed on human dendritic cells (DCs), and thus the latter cell type also receives signals via cell-bound IgE. Notably, steady-state expression of FcɛRI on DCs is absent in SPF-housed mice. How DCs integrate IgE/FcɛRI-derived signals into their sentinel functions as gatekeepers of immunity was therefore only recently studied with transgenic mice that phenocopy human FcɛRI expression. In this review, we summarize advances in our understanding of the functions of DC-bound IgE which demonstrate that IgE-mediated activation of DCs in allergic Th2-type inflammation appears to be immune regulatory rather than pro-inflammatory.


Asunto(s)
Células Dendríticas/inmunología , Hipersensibilidad/inmunología , Inmunoglobulina E/inmunología , Receptores de IgE/inmunología , Animales , Presentación de Antígeno , Basófilos/inmunología , Basófilos/patología , Degranulación de la Célula , Células Dendríticas/patología , Expresión Génica , Humanos , Hipersensibilidad/genética , Hipersensibilidad/patología , Inmunoglobulina E/genética , Mastocitos/inmunología , Mastocitos/patología , Ratones , Ratones Transgénicos , Unión Proteica , Receptores de IgE/genética , Células Th2/inmunología , Células Th2/patología
11.
J Exp Med ; 212(4): 457-67, 2015 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-25824821

RESUMEN

Immunoglobulin ε (IgE) antibodies are the primary mediators of allergic diseases, which affect more than 1 in 10 individuals worldwide. IgE specific for innocuous environmental antigens, or allergens, binds and sensitizes tissue-resident mast cells expressing the high-affinity IgE receptor, FcεRI. Subsequent allergen exposure cross-links mast cell-bound IgE, resulting in the release of inflammatory mediators and initiation of the allergic cascade. It is well established that precise glycosylation patterns exert profound effects on the biological activity of IgG. However, the contribution of glycosylation to IgE biology is less clear. Here, we demonstrate an absolute requirement for IgE glycosylation in allergic reactions. The obligatory glycan was mapped to a single N-linked oligomannose structure in the constant domain 3 (Cε3) of IgE, at asparagine-394 (N394) in human IgE and N384 in mouse. Genetic disruption of the site or enzymatic removal of the oligomannose glycan altered IgE secondary structure and abrogated IgE binding to FcεRI, rendering IgE incapable of eliciting mast cell degranulation, thereby preventing anaphylaxis. These results underscore an unappreciated and essential requirement of glycosylation in IgE biology.


Asunto(s)
Anafilaxia/inmunología , Degranulación de la Célula/inmunología , Inmunoglobulina E/inmunología , Mastocitos/inmunología , Polisacáridos/inmunología , Receptores de IgE/inmunología , Anafilaxia/genética , Anafilaxia/patología , Anafilaxia/prevención & control , Animales , Degranulación de la Célula/genética , Glicosilación , Humanos , Regiones Constantes de Inmunoglobulina/genética , Regiones Constantes de Inmunoglobulina/inmunología , Inmunoglobulina E/genética , Mediadores de Inflamación , Mastocitos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Polisacáridos/genética , Receptores de IgE/genética
12.
Cell Rep ; 10(9): 1487-1495, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25753415

RESUMEN

Epidemiologic studies discovered an inverse association between immunoglobulin E (IgE)-mediated allergies and cancer, implying tumor-protective properties of IgE. However, the underlying immunologic mechanisms remain poorly understood. Antigen cross-presentation by dendritic cells (DCs) is of key importance for anti-tumor immunity because it induces the generation of cytotoxic CD8+ T lymphocytes (CTLs) with specificity for tumor antigens. We demonstrate that DCs use IgE and FcεRI, the high-affinity IgE receptor, for cross-presentation and priming of CTLs in response to free soluble antigen at low doses. Importantly, IgE/FcεRI-mediated cross-presentation is a distinct receptor-mediated pathway because it does not require MyD88 signals or IL-12 induction in DCs. Using passive immunization with tumor antigen-specific IgE and DC-based vaccination experiments, we demonstrate that IgE-mediated cross-presentation significantly improves anti-tumor immunity and induces memory responses in vivo. Our findings suggest a cellular mechanism for the tumor-protective features of IgE and expand the known physiological functions of this immunoglobulin.

13.
Front Immunol ; 5: 140, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24744762

RESUMEN

The ability of dendritic cells (DCs) to cross-present tumor antigens has long been a focus of interest to physicians, as well as basic scientists, that aim to establish efficient cell-based cancer immune therapy. A prerequisite for exploiting this pathway for therapeutic purposes is a better understanding of the mechanisms that underlie the induction of tumor-specific cytotoxic T-lymphocyte (CTL) responses when initiated by DCs via cross-presentation. The ability of humans DC to perform cross-presentation is of utmost interest, as this cell type is a main target for cell-based immunotherapy in humans. The outcome of a cross-presentation event is guided by the nature of the antigen, the form of antigen uptake, and the subpopulation of DCs that performs presentation. Generally, CD8α(+) DCs are considered to be the most potent cross-presenting DCs. This paradigm, however, only applies to soluble antigens. During adaptive immune responses, immune complexes form when antibodies interact with their specific epitopes on soluble antigens. Immunoglobulin G (IgG) immune complexes target Fc-gamma receptors on DCs to shuttle exogenous antigens efficiently into the cross-presentation pathway. This receptor-mediated cross-presentation pathway is a well-described route for the induction of strong CD8(+) T cell responses. IgG-mediated cross-presentation is intriguing because it permits the CD8(-) DCs, which are commonly considered to be weak cross-presenters, to efficiently cross-present. Engaging multiple DC subtypes for cross-presentation might be a superior strategy to boost CTL responses in vivo. We here summarize our current understanding of how DCs use IgG-complexed antigens for the efficient induction of CTL responses. Because of its importance for human cell therapy, we also review the recent advances in the characterization of cross-presentation properties of human DC subsets.

14.
Cell Host Microbe ; 13(5): 558-569, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23684307

RESUMEN

The plasma membrane and all membrane-bound organelles except for the Golgi and endoplasmic reticulum (ER) are equipped with pattern-recognition molecules to sense microbes or their products and induce innate immunity for host defense. Here, we report that inositol-requiring-1α (IRE1α), an ER protein that signals in the unfolded protein response (UPR), is activated to induce inflammation by binding a portion of cholera toxin as it co-opts the ER to cause disease. Other known UPR transducers, including the IRE1α-dependent transcription factor XBP1, are dispensable for this signaling. The inflammatory response depends instead on the RNase activity of IRE1α to degrade endogenous mRNA, a process termed regulated IRE1α-dependent decay (RIDD) of mRNA. The mRNA fragments produced engage retinoic-acid inducible gene 1 (RIG-I), a cytosolic sensor of RNA viruses, to activate NF-κB and interferon pathways. We propose IRE1α provides for a generalized mechanism of innate immune surveillance originating within the ER lumen.


Asunto(s)
Toxina del Cólera/inmunología , Toxina del Cólera/metabolismo , ARN Helicasas DEAD-box/inmunología , Endorribonucleasas/inmunología , Endorribonucleasas/metabolismo , Inmunidad Innata , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Línea Celular , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/metabolismo , Humanos , Unión Proteica , Receptores Inmunológicos
16.
Immunol Lett ; 141(1): 36-44, 2011 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-21920387

RESUMEN

Soluble isoforms of three human IgE Fc receptors, namely FcεRI, FcεRII, and galectin-3, can be found in serum. These soluble IgE receptors are a diverse family of proteins unified by the characteristic of interacting with IgE in the extracellular matrix. A truncated form of the alpha-chain of FcεRI, the high affinity IgE receptor, has recently been described as a soluble isoform (sFcεRI). Multiple soluble isoforms of CD23 (sCD23), the low affinity IgE receptor also known as FcεRII, are generated via different mechanisms of extracellular and intracellular proteolysis. The second low affinity IgE receptor, galectin-3, only exists as a secretory protein. We here discuss the physiological roles of these three soluble IgE receptors as elements of the human IgE network. Additionally, we review the potential and current use of sFcεRI, sCD23, and galectin-3 as biomarkers in human disease.


Asunto(s)
Galectina 3 , Inmunoglobulina E/metabolismo , Receptores de IgE , Biomarcadores/sangre , Galectina 3/química , Galectina 3/inmunología , Galectina 3/metabolismo , Humanos , Inmunoglobulina E/inmunología , Unión Proteica/fisiología , Mapas de Interacción de Proteínas/fisiología , Isoformas de Proteínas/sangre , Isoformas de Proteínas/inmunología , Proteolisis , Receptores de IgE/química , Receptores de IgE/inmunología , Receptores de IgE/metabolismo
17.
J Immunol Methods ; 373(1-2): 192-9, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21903095

RESUMEN

The aim of this study was to develop a standardized enzyme-linked immunosorbent assay (ELISA) for detection of human soluble Fc-epsilon-RI (sFcεRI), a serum isoform of the high affinity IgE receptor. A recombinant version of sFcεRI was produced in baculovirus and used as standard. ELISA plates were coated with anti-mouse IgG followed by incubation with the monoclonal capture antibody CRA1. This FcεRI-alpha-specific antibody binds to the stalk region of the protein and does not inhibit IgE-binding. After incubation with standards or serum samples, plates were incubated with chimeric IgE followed by detection with horseradish peroxidase conjugated anti-human IgE. Enzymatic activity was visualized with (3,3',5,5')-tetramethylbenzidine. Specificity was demonstrated by omission of capture or detection reagents. Units (U) of detection were established and the dynamic range of the assay was defined as 10-640 U/ml for a 1/5 serum dilution. Parameters of linearity (R(2)>0.999), matrix interference test (recovery of 70-110%), intra-assay variability (coefficient of variation (CV) <20%) and inter-assay variability (CV <20%) met acceptance criteria for immunoassay validation. Correlation analysis of serum units of sFcεRI measured with the new ELISA and serum IgE levels confirmed earlier published data describing a weak correlation of the two parameters in patients with elevated serum IgE while no correlation in patients with normal serum IgE or the total patient group was found. In summary, we established and validated a standardized ELISA for the detection of sFcεRI. This novel method now allows for comparative analysis of sFcεRI levels in health and disease.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Ensayo de Inmunoadsorción Enzimática/métodos , Receptores de IgE/sangre , Receptores de IgE/inmunología , Adolescente , Afinidad de Anticuerpos/inmunología , Niño , Preescolar , Humanos , Immunoblotting , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Lactante , Receptores de IgE/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/normas , Estándares de Referencia , Valores de Referencia , Reproducibilidad de los Resultados
18.
PLoS One ; 6(4): e19098, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21544204

RESUMEN

Soluble IgE receptors are potential in vivo modulators of IgE-mediated immune responses and are thus important for our basic understanding of allergic responses. We here characterize a novel soluble version of the IgE-binding alpha-chain of Fc-epsilon-RI (sFcεRI), the high affinity receptor for IgE. sFcεRI immunoprecipitates as a protein of ∼40 kDa and contains an intact IgE-binding site. In human serum, sFcεRI is found as a soluble free IgE receptor as well as a complex with IgE. Using a newly established ELISA, we show that serum sFcεRI levels correlate with serum IgE in patients with elevated IgE. We also show that serum of individuals with normal IgE levels can be found to contain high levels of sFcεRI. After IgE-antigen-mediated crosslinking of surface FcεRI, we detect sFcεRI in the exosome-depleted, soluble fraction of cell culture supernatants. We further show that sFcεRI can block binding of IgE to FcεRI expressed at the cell surface. In summary, we here describe the alpha-chain of FcεRI as a circulating soluble IgE receptor isoform in human serum.


Asunto(s)
Receptores de IgE/sangre , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunoglobulina E/metabolismo , Unión Proteica/genética , Isoformas de Proteínas/sangre , Isoformas de Proteínas/metabolismo , Receptores de IgE/metabolismo
19.
Mol Immunol ; 46(11-12): 2333-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19406478

RESUMEN

The family of activating immune receptors stabilizes via the 3-helix assembly principle. A charged basic transmembrane residue interacts with two charged acidic transmembrane residues and forms a 3-helix interface to stabilize receptor complexes in the lipid bilayer. One family member, the high affinity receptor for IgE, Fc epsilon RI, is a key regulator of immediate allergic responses. Tetrameric Fc epsilon RI consists of the IgE-binding alpha-chain, the multimembrane-spanning beta-chain and a dimer of the gamma-subunit (Fc epsilon R gamma). Comparative analysis of these seven transmembrane regions indicates that Fc epsilon RI does not meet the charge requirements for the 3-helix assembly mechanism. We performed alanine mutagenesis to show that the only basic amino acid in the transmembrane regions, beta K97, is not involved in Fc epsilon RI stabilization or surface upregulation, a hallmark function of the beta-chain. Even a beta K97E mutant is functional despite four negatively charged acidic amino acids in the transmembrane regions. Using truncation mutants, we demonstrate that the first uncharged transmembrane domain of the beta-chain contains the interface for receptor stabilization. In vitro translation experiments depict the first transmembrane region as the internal signal peptide of the beta-chain. We also show that this beta-chain domain can function as a cleavable signal peptide when used as a leader peptide for a Type I protein. Our results provide evidence that tetrameric Fc epsilon RI does not assemble according to the 3-helix assembly principle. We conclude that receptors formed with multispanning proteins use different mechanisms of shielding transmembrane charged amino acids.


Asunto(s)
Membrana Dobles de Lípidos/química , Receptores de IgE/química , Secuencia de Aminoácidos , Línea Celular , Humanos , Datos de Secuencia Molecular , Mutación , Conformación Proteica , Multimerización de Proteína , Estabilidad Proteica , Estructura Terciaria de Proteína , Receptores de IgE/biosíntesis , Receptores de IgE/genética
20.
Blood ; 109(3): 971-9, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17018855

RESUMEN

Neutrophil granulocytes (Gs) represent highly abundant and short-lived leukocytes that are constantly regenerated from a small pool of myeloid committed progenitors. Nuclear receptor (NR) family members are ligand-activated transcription factors that play key roles in cellular proliferation and differentiation processes including myelopoiesis. Retinoid X receptor alpha (RXRalpha) represents the predominant NR types I and II homo- and heterodimerization partner in myeloid cells. Here we show that human myeloid progenitors express RXRalpha protein at sustained high levels during macrophage colony-stimulating factor (M-CSF)-induced monopoiesis. In sharp contrast, RXRalpha is down-regulated during G-CSF-dependent late-stage neutrophil differentiation from myeloid progenitors. Down-regulation of RXRalpha is critically required for neutrophil development since ectopic RXRalpha inhibited granulopoiesis by impairing proliferation and differentiation. Moreover, ectopic RXRalpha was sufficient to redirect G-CSF-dependent granulocyte differentiation to the monocyte lineage and to promote M-CSF-induced monopoiesis. Functional genetic interference with RXRalpha signaling in hematopoietic progenitor/stem cells using a dominant-negative RXRalpha promoted the generation of late-stage granulocytes in human cultures in vitro and in reconstituted mice in vivo. Therefore, our data suggest that RXRalpha down-regulation is a critical requirement for the generation of neutrophil granulocytes.


Asunto(s)
Células Progenitoras Mieloides/citología , Neutrófilos/citología , Receptor alfa X Retinoide/genética , Animales , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Regulación hacia Abajo/genética , Humanos , Factor Estimulante de Colonias de Macrófagos/farmacología , Ratones , Mielopoyesis/efectos de los fármacos , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA