Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Environ Microbiol ; 24(12): 5924-5935, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35799468

RESUMO

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.


Assuntos
Chlorella , Cilióforos , Paramecium , Simbiose , Análise Custo-Benefício
2.
Cancer Res ; 81(11): 3079-3091, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33504557

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Neoplasias do Colo/tratamento farmacológico , Regulação Neoplásica da Expressão Gênica , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Células Estromais/imunologia , Microambiente Tumoral/imunologia , Proteína Supressora de Tumor p53/antagonistas & inibidores , Animais , Apoptose , Proliferação de Células , Neoplasias do Colo/imunologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Quimioterapia Combinada , Feminino , Humanos , Imidazóis/farmacologia , Inibidores de Checkpoint Imunológico/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Nus , Pirimidinas/farmacologia , Pirróis/farmacologia , Células Estromais/efeitos dos fármacos , Células Tumorais Cultivadas , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Mol Immunol ; 68(2 Pt A): 116-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26052071

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Hipersensibilidade/imunologia , Imunoglobulina E/imunologia , Receptores de IgE/imunologia , Animais , Apresentação de Antígeno , Basófilos/imunologia , Basófilos/patologia , Degranulação Celular , Células Dendríticas/patologia , Expressão Gênica , Humanos , Hipersensibilidade/genética , Hipersensibilidade/patologia , Imunoglobulina E/genética , Mastócitos/imunologia , Mastócitos/patologia , Camundongos , Camundongos Transgênicos , Ligação Proteica , Receptores de IgE/genética , Células Th2/imunologia , Células Th2/patologia
5.
J Exp Med ; 212(4): 457-67, 2015 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-25824821

RESUMO

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.


Assuntos
Anafilaxia/imunologia , Degranulação Celular/imunologia , Imunoglobulina E/imunologia , Mastócitos/imunologia , Polissacarídeos/imunologia , Receptores de IgE/imunologia , Anafilaxia/genética , Anafilaxia/patologia , Anafilaxia/prevenção & controle , Animais , Degranulação Celular/genética , Glicosilação , Humanos , Regiões Constantes de Imunoglobulina/genética , Regiões Constantes de Imunoglobulina/imunologia , Imunoglobulina E/genética , Mediadores da Inflamação , Mastócitos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Polissacarídeos/genética , Receptores de IgE/genética
6.
Cell Rep ; 10(9): 1487-1495, 2015 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-25753415

RESUMO

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.

7.
Front Immunol ; 5: 140, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24744762

RESUMO

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.

8.
Cell Host Microbe ; 13(5): 558-569, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23684307

RESUMO

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.


Assuntos
Toxina da Cólera/imunologia , Toxina da Cólera/metabolismo , RNA Helicases DEAD-box/imunologia , Endorribonucleases/imunologia , Endorribonucleases/metabolismo , Imunidade Inata , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Linhagem Celular , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Humanos , Ligação Proteica , Receptores Imunológicos
9.
Cancer Immunol Immunother ; 61(9): 1521-5, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22042251

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Imunoglobulina E/imunologia , Receptores de IgG/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Apresentação de Antígeno/imunologia , Apresentação Cruzada/imunologia , Células Dendríticas/metabolismo , Humanos , Imunoglobulina E/metabolismo , Receptores de IgG/metabolismo , Linfócitos T Citotóxicos/metabolismo
10.
Immunol Lett ; 141(1): 36-44, 2011 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-21920387

RESUMO

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.


Assuntos
Galectina 3 , Imunoglobulina E/metabolismo , Receptores de IgE , Biomarcadores/sangue , Galectina 3/química , Galectina 3/imunologia , Galectina 3/metabolismo , Humanos , Imunoglobulina E/imunologia , Ligação Proteica/fisiologia , Mapas de Interação de Proteínas/fisiologia , Isoformas de Proteínas/sangue , Isoformas de Proteínas/imunologia , Proteólise , Receptores de IgE/química , Receptores de IgE/imunologia , Receptores de IgE/metabolismo
11.
J Immunol Methods ; 373(1-2): 192-9, 2011 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-21903095

RESUMO

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.


Assuntos
Anticorpos Monoclonais/imunologia , Ensaio de Imunoadsorção Enzimática/métodos , Receptores de IgE/sangue , Receptores de IgE/imunologia , Adolescente , Afinidade de Anticorpos/imunologia , Criança , Pré-Escolar , Humanos , Immunoblotting , Imunoglobulina E/sangue , Imunoglobulina E/imunologia , Lactente , Receptores de IgE/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/normas , Padrões de Referência , Valores de Referência , Reprodutibilidade dos Testes
12.
Proc Natl Acad Sci U S A ; 108(24): 9927-32, 2011 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-21628593

RESUMO

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.


Assuntos
Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Imunoglobulina G/imunologia , Receptores Fc/imunologia , Animais , Antígenos/imunologia , Western Blotting , Antígeno CD11b/imunologia , Antígeno CD11b/metabolismo , Antígenos CD8/imunologia , Antígenos CD8/metabolismo , Colite/induzido quimicamente , Colite/imunologia , Colite/metabolismo , Citosol/imunologia , Citosol/metabolismo , Células Dendríticas/metabolismo , Sulfato de Dextrana , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Concentração de Íons de Hidrogênio , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mutação , NADPH Oxidase 2 , NADPH Oxidases/imunologia , NADPH Oxidases/metabolismo , Fagossomos/imunologia , Fagossomos/metabolismo , Ligação Proteica , Receptores Fc/genética , Receptores Fc/metabolismo , Receptores de IgG/imunologia , Receptores de IgG/metabolismo , ATPases Vacuolares Próton-Translocadoras/imunologia , ATPases Vacuolares Próton-Translocadoras/metabolismo , Proteínas rab de Ligação ao GTP/imunologia , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab27 de Ligação ao GTP
13.
J Immunol ; 187(1): 164-71, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21622859

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/patologia , Mediadores da Inflamação/metabolismo , Receptores de IgE/metabolismo , Células Th2/imunologia , Células Th2/patologia , Alérgenos/toxicidade , Animais , Antígenos de Plantas/toxicidade , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/metabolismo , Feminino , Humanos , Mediadores da Inflamação/fisiologia , Mediadores da Inflamação/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ovalbumina/toxicidade , Ligação Proteica/genética , Ligação Proteica/imunologia , Receptores de IgE/deficiência , Receptores de IgE/fisiologia , Células Th2/metabolismo , Fatores de Tempo
14.
PLoS One ; 6(4): e19098, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21544204

RESUMO

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.


Assuntos
Receptores de IgE/sangue , Ensaio de Imunoadsorção Enzimática , Humanos , Imunoglobulina E/metabolismo , Ligação Proteica/genética , Isoformas de Proteínas/sangue , Isoformas de Proteínas/metabolismo , Receptores de IgE/metabolismo
15.
J Immunol ; 185(6): 3217-26, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20733204

RESUMO

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.


Assuntos
Sistema y+ de Transporte de Aminoácidos/fisiologia , Apresentação de Antígeno/imunologia , Diferenciação Celular/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Sistema y+ de Transporte de Aminoácidos/antagonistas & inibidores , Animais , Apresentação de Antígeno/genética , Transporte Biológico/imunologia , Diferenciação Celular/genética , Células Cultivadas , Apresentação Cruzada/genética , Apresentação Cruzada/imunologia , Cistina/metabolismo , Células Dendríticas/metabolismo , Ácido Glutâmico/metabolismo , Glutationa/antagonistas & inibidores , Glutationa/metabolismo , Homeostase/imunologia , Humanos , Líquido Intracelular/imunologia , Líquido Intracelular/metabolismo , Lipopolissacarídeos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovalbumina/imunologia , Ovalbumina/metabolismo
16.
J Biol Chem ; 285(20): 15314-15323, 2010 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-20304923

RESUMO

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.


Assuntos
Sinais Direcionadores de Proteínas , Receptores de IgE/metabolismo , Tirosina/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Sequência de Bases , Linhagem Celular , Membrana Celular/metabolismo , Primers do DNA , Retículo Endoplasmático/metabolismo , Citometria de Fluxo , Complexo de Golgi/metabolismo , Humanos , Microscopia de Fluorescência , Dados de Sequência Molecular , Transporte Proteico , Receptores de IgE/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
J Immunol ; 183(1): 66-74, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19535631

RESUMO

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.


Assuntos
Diferenciação Celular/imunologia , Células de Langerhans/imunologia , Células de Langerhans/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem da Célula/efeitos dos fármacos , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Inibidores do Crescimento/farmacologia , Inibidores do Crescimento/fisiologia , Humanos , Células K562 , Células de Langerhans/citologia , Monócitos/citologia , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/imunologia , Células Progenitoras Mieloides/metabolismo , Pirimidinas/farmacologia , Receptores de Hidrocarboneto Arílico/agonistas , Receptores de Hidrocarboneto Arílico/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/imunologia
18.
Mol Immunol ; 46(11-12): 2333-9, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19406478

RESUMO

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.


Assuntos
Bicamadas Lipídicas/química , Receptores de IgE/química , Sequência de Aminoácidos , Linhagem Celular , Humanos , Dados de Sequência Molecular , Mutação , Conformação Proteica , Multimerização Proteica , Estabilidade Proteica , Estrutura Terciária de Proteína , Receptores de IgE/biossíntese , Receptores de IgE/genética
19.
Blood ; 109(3): 971-9, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17018855

RESUMO

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.


Assuntos
Células Progenitoras Mieloides/citologia , Neutrófilos/citologia , Receptor X Retinoide alfa/genética , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células , Células Cultivadas , Regulação para Baixo/genética , Humanos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Camundongos , Mielopoese/efeitos dos fármacos , Transdução Genética
20.
Blood ; 109(1): 185-93, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16960152

RESUMO

Environmentally exposed epithelial Langerhans cells (LCs) encounter diverse innate stress signals, which lead to the activation of complex intracellular signaling cascades. Among these, p38 MAPK is consistently phosphorylated. For which aspects of LC activation triggering of p38 signaling is sufficient remains to be elucidated. We show that conditional induction of a dominant active form of MAPK kinase 6 (d.a.MKK6), a direct upstream kinase of p38, in LCs efficiently induces the up-regulation of costimulatory molecules and enhances their T-cell stimulatory capacity. These immediate effects showed no or only a minor requirement for classical NF-kappaB signaling. Concomitant with LC activation, d.a.MKK6 induced the alternative NF-kappaB member RelB, whose nuclear localization marks mature DCs. Specific inhibition of nuclear RelB during d.a.MKK6-induced LC activation further enhanced their maturation state. This observation was validated using the p38 activator anisomycin, thus suggesting a novel LC intrinsic control mechanism regulated by RelB.


Assuntos
Células de Langerhans/citologia , MAP Quinase Quinase 6/fisiologia , Fator de Transcrição RelB/fisiologia , Anisomicina/farmacologia , Apresentação de Antígeno , Antígeno B7-1/biossíntese , Antígeno B7-1/genética , Antígeno B7-2/biossíntese , Antígeno B7-2/genética , Caspase 1/metabolismo , Diferenciação Celular , Núcleo Celular/metabolismo , Células Cultivadas/citologia , Células Cultivadas/metabolismo , Meios de Cultura Livres de Soro , Citocinas/farmacologia , Doxiciclina/farmacologia , Ativação Enzimática , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Genes Dominantes , Humanos , Interleucina-1beta/biossíntese , Interleucina-1beta/genética , Células de Langerhans/metabolismo , MAP Quinase Quinase 6/genética , Modelos Imunológicos , NF-kappa B/fisiologia , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Receptores de Fator de Crescimento Neural/biossíntese , Receptores de Fator de Crescimento Neural/genética , Linfócitos T/imunologia , Células U937 , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...