Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Biomedicines ; 10(5)2022 May 17.
Article in English | MEDLINE | ID: mdl-35625889

ABSTRACT

BACKGROUND: Pathogens or trauma-derived danger signals induced maturation and activation of plasmacytoid dendritic cells (pDCs) is a pivotal step in pDC-dependent host defense. Exposure of pDC to cardiometabolic disease-associated lipids and proteins may well influence critical signaling pathways, thereby compromising immune responses against endogenous, bacterial and viral pathogens. In this study, we have addressed if hyperlipidemia impacts human pDC activation, cytokine response and capacity to prime CD4+ T cells. METHODS AND RESULTS: We show that exposure to pro-atherogenic oxidized low-density lipoproteins (oxLDL) led to pDC lipid accumulation, which in turn ablated a Toll-like receptor (TLR) 7 and 9 dependent up-regulation of pDC maturation markers CD40, CD83, CD86 and HLA-DR. Moreover, oxLDL dampened TLR9 activation induced the production of pro-inflammatory cytokines in a NUR77/IRF7 dependent manner and impaired the capacity of pDCs to prime and polarize CD4+ T helper (Th) cells. CONCLUSION: Our findings reveal profound effects of dyslipidemia on pDC responses to pathogen-derived signals.

2.
BMC Immunol ; 19(1): 8, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29433450

ABSTRACT

BACKGROUND: Besides their prominent role in the elimination of infected or malignantly transformed cells, natural killer (NK) cells serve as modulators of adaptive immune responses. Enhancing bidirectional crosstalk between NK cells and dendritic cells (DC) is considered a promising tool to potentiate cancer vaccines. We investigated to what extent direct sensing of viral and bacterial motifs by NK cells contributes to the response of inflammatory DC against the same pathogenic stimulus. RESULTS: We demonstrated that sensing of bacterial and viral PAMPs by NK cells contributes to DC cytokine production via NK cell-derived soluble factors. This enhancement of DC cytokine production was dependent on the pattern recognition receptor (PRR) agonist but also on the cytokine environment in which NK cells recognized the pathogen, indicating the importance of accessory cell activation for this mechanism. We showed in blocking experiments that NK cell-mediated amplification of DC cytokine secretion is dependent on NK cell-derived IFN-γ irrespective of the PRR that is sensed by the NK cell. CONCLUSIONS: These findings illustrate the importance of bidirectional interaction between different PRR-expressing immune cells, which can have implications on the selection of adjuvants for vaccination strategies.


Subject(s)
Cytokines/immunology , Dendritic Cells/immunology , Inflammation Mediators/immunology , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Monocytes/immunology , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/metabolism , Humans , Inflammation Mediators/metabolism , Interferon-gamma/metabolism , Killer Cells, Natural/microbiology , Killer Cells, Natural/virology , Lymphocyte Activation/immunology , Monocytes/metabolism , Pathogen-Associated Molecular Pattern Molecules/immunology , Pathogen-Associated Molecular Pattern Molecules/metabolism , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism
3.
PLoS One ; 11(7): e0159515, 2016.
Article in English | MEDLINE | ID: mdl-27427766

ABSTRACT

Accumulating evidence indicates that fractionated radiotherapy (RT) can result in distant non-irradiated (abscopal) tumour regression. Although preclinical studies indicate the importance of T cells in this infrequent phenomenon, these studies do not preclude that other immune mechanisms exhibit an addition role in the abscopal effect. We therefore addressed the question whether in addition to T cell mediated responses also humoral anti-tumour responses are modulated after fractionated RT and whether systemic dendritic cell (DC) stimulation can enhance tumour-specific antibody production. We selected the 67NR mammary carcinoma model since this tumour showed spontaneous antibody production in all tumour-bearing mice. Fractionated RT to the primary tumour was associated with a survival benefit and a delayed growth of a non-irradiated (contralateral) secondary tumour. Notably, fractionated RT did not affect anti-tumour antibody titers and the composition of the immunoglobulin (Ig) isotypes. Likewise, we demonstrated that treatment of tumour-bearing Balb/C mice with DC stimulating growth factor Flt3-L did neither modulate the magnitude nor the composition of the humoral immune response. Finally, we evaluated the immune infiltrate and Ig isotype content of the tumour tissue using flow cytometry and found no differences between treatment groups that were indicative for local antibody production. In conclusion, we demonstrate that the 67NR mammary carcinoma in Balb/C mice is associated with a pre-existing antibody response. And, we show that in tumour-bearing Balb/C mice with abscopal tumour regression such pre-existing antibody responses are not altered upon fractionated RT and/or DC stimulation with Flt3-L. Our research indicates that evaluating the humoral immune response in the setting of abscopal tumour regression is not invariably associated with therapeutic effects.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Carcinoma/radiotherapy , Gamma Rays/therapeutic use , Immunity, Humoral , Mammary Glands, Animal/radiation effects , Mammary Neoplasms, Experimental/radiotherapy , Animals , Carcinoma/immunology , Carcinoma/pathology , Dendritic Cells/immunology , Dendritic Cells/pathology , Dose Fractionation, Radiation , Female , Immunoglobulin Isotypes/blood , Mammary Glands, Animal/immunology , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Membrane Proteins/pharmacology , Mice , Mice, Inbred BALB C , T-Lymphocytes/immunology , T-Lymphocytes/pathology
4.
Mediators Inflamm ; 2016: 5740373, 2016.
Article in English | MEDLINE | ID: mdl-26980946

ABSTRACT

A coordinated cellular interplay is of crucial importance in both host defense against pathogens and malignantly transformed cells. The various interactions of Dendritic Cells (DC), Natural Killer (NK) cells, and T helper (Th) cells can be influenced by a variety of pathogen-associated molecular patterns (PAMPs) and will lead to enhanced CD8(+) effector T cell responses. Specific Pattern Recognition Receptor (PRR) triggering during maturation enables DC to enhance Th1 as well as NK helper cell responses. This effect is correlated with the amount of IL-12p70 released by DC. Activated NK cells are able to amplify the proinflammatory cytokine profile of DC via the release of IFN-γ. The knowledge on how PAMP recognition can modulate the DC is of importance for the design and definition of appropriate therapeutic cancer vaccines. In this review we will discuss the potential role of specific PAMP-matured DC in optimizing therapeutic DC-based vaccines, as some of these DC are efficiently activating Th1, NK cells, and cytotoxic T cells. Moreover, to optimize these vaccines, also the inhibitory effects of tumor-derived suppressive factors, for example, on the NK-DC crosstalk, should be taken into account. Finally, the suppressive role of the tumor microenvironment in vaccination efficacy and some proposals to overcome this by using combination therapies will be described.


Subject(s)
Dendritic Cells/metabolism , Killer Cells, Natural/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Dendritic Cells/immunology , Humans , Killer Cells, Natural/immunology , Models, Biological , T-Lymphocytes, Helper-Inducer/immunology
5.
J Interferon Cytokine Res ; 35(9): 748-58, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26134473

ABSTRACT

Besides T helper (Th) cells, natural killer (NK) cells have also been described to participate in the shaping of dendritic cell (DC)-mediated adaptive immune responses. At present, it remains unclear to what extent the induction of these NK helper cell immune mechanisms is coupled with Th responses and whether both helper immune responses are induced by the same DC upon specific pathogen recognition receptor (PRR) stimulation. In this study, we demonstrate that maturation of DCs with a cocktail containing FMKp (membrane fragments of Klebsiella pneumoniae) mounts both Th cell and NK cell helper responses in a PRR-triggered dose-dependent manner as determined by the capacity of the helper cells to produce IFN-γ. Furthermore, by triggering an additional PRR pathway [FMKp in combination with poly(I:C) lyovec], we reveal that both approaches modulate the amount of DC-derived IL-12p70 and that this cytokine is the key determinant of the DC-induced Th1 and NK cell helper responses. Moreover, all PRR triggers able to induce IL-12-producing mature DCs are sufficient to induce these helper responses. We propose the existence of a single program used by DCs to induce potent cellular immune responses by stimulating both T helper and NK cell helper processes. This knowledge can help to select the proper PRR triggers in preventive and therapeutic vaccine design.


Subject(s)
Dendritic Cells/immunology , Interleukin-12/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Membranes/immunology , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/immunology , Cells, Cultured , Coculture Techniques/methods , Cytokines/immunology , Cytotoxicity, Immunologic/immunology , Humans , Interferon-gamma/immunology , Klebsiella pneumoniae/immunology , Poly I-C/immunology , Signal Transduction/immunology
6.
PLoS One ; 9(8): e103725, 2014.
Article in English | MEDLINE | ID: mdl-25144736

ABSTRACT

A crucial step in generating de novo immune responses is the polarization of naive cognate CD4+ T cells by pathogen-triggered dendritic cells (DC). In the human setting, standardized DC-dependent systems are lacking to study molecular events during the initiation of a naive CD4+ T cell response. We developed a TCR-restricted assay to compare different pathogen-triggered human DC for their capacities to instruct functional differentiation of autologous, naive CD4+ T cells. We demonstrated that this methodology can be applied to compare differently matured DC in terms of kinetics, direction, and magnitude of the naive CD4+ T cell response. Furthermore, we showed the applicability of this assay to study the T cell polarizing capacity of low-frequency blood-derived DC populations directly isolated ex vivo. This methodology for addressing APC-dependent instruction of naive CD4+ T cells in a human autologous setting will provide researchers with a valuable tool to gain more insight into molecular mechanisms occurring in the early phase of T cell polarization. In addition, it may also allow the study of pharmacological agents on DC-dependent T cell polarization in the human system.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Dendritic Cells/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/physiology , Cell Polarity/physiology , Cells, Cultured , Dendritic Cells/metabolism , Flow Cytometry , Humans , Kinetics , Monocytes/cytology , Polymerase Chain Reaction
7.
Clin Cancer Res ; 20(5): 1095-103, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24590885

ABSTRACT

In cancer therapy, dendritic cell (DC) vaccination is still being explored. Clinical responses, however, are diverse and there is a lack of immunologic readout systems that correspond with clinical outcome. Only in the minority of patients, T-cell responses correlate with clinical outcome, indicating that other immune cells also gain anticancer activity. We still have limited knowledge of the effect of DC vaccination on different immune effector cells. However, it has been shown that bidirectional cross-talk between natural killer (NK) cells and DCs is responsible for enhanced activation of both cell types and increases their antitumor activity. In this review, we postulate the possibility that NK cells are the secret weapons in DC vaccination and studying their behavior together with T-cell activation in vaccinated individuals might predict clinical outcome.


Subject(s)
Killer Cells, Natural/immunology , Neoplasms/immunology , Neoplasms/therapy , Animals , Cancer Vaccines/immunology , Cell Communication/immunology , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immunotherapy , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , Neoplasms/metabolism
8.
Blood ; 118(9): 2473-82, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21715307

ABSTRACT

Among prostaglandins (PGs), PGE2 is abundantly expressed in various malignancies and is probably one of many factors promoting tumor growth by inhibiting tumor immune surveillance. In the current study, we report on a novel mechanism by which PGE2 inhibits in vitro natural killer-dendritic cell (NK-DC) crosstalk and thereby innate and adaptive immune responses via its effect on NK-DC crosstalk. The presence of PGE2 during IFN-γ/membrane fraction of Klebsiella pneumoniae DC maturation inhibits the production of chemokines (CCL5, CCL19, and CXCL10) and cytokines (IL-12 and IL-18), which is cAMP-dependent and imprinted during DC maturation. As a consequence, these DCs fail to attract NK cells and show a decreased capacity to trigger NK cell IFN-γ production, which in turn leads to reduced T-helper 1 polarization. In addition, the presence of PGE2 during DC maturation impairs DC-mediated augmentation of NK-cell cytotoxicity. Opposed to their inhibitory effects on peripheral blood-derived NK cells, PGE2 matured DCs induce IL-22 secretion of inflammation constraining NKp44(+) NK cells present in mucosa-associated lymphoid tissue. The inhibition of NK-DC interaction is a novel regulatory property of PGE2 that is of possible relevance in dampening immune responses in vivo.


Subject(s)
Dendritic Cells/drug effects , Dinoprostone/pharmacology , Inflammation/immunology , Killer Cells, Natural/drug effects , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Bucladesine/pharmacology , Cell Differentiation , Cell Movement/drug effects , Cells, Cultured/drug effects , Cells, Cultured/immunology , Chemokines/biosynthesis , Chemokines/genetics , Coculture Techniques , Cytokines/biosynthesis , Cytokines/genetics , Cytotoxicity, Immunologic/drug effects , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Down-Regulation/drug effects , Gene Expression Regulation/drug effects , Humans , Immunosuppression Therapy , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Killer Cells, Natural/immunology , Klebsiella pneumoniae/immunology , Misoprostol/pharmacology , Palatine Tonsil/cytology , T-Lymphocytes, Helper-Inducer/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...