Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Eur J Immunol ; 48(11): 1796-1809, 2018 11.
Article in English | MEDLINE | ID: mdl-30184252

ABSTRACT

Type I and type III interferons (IFNs) are fundamental for antiviral immunity, but prolonged expression is also detrimental to the host. Therefore, upon viral infection high levels of type I and III IFNs are followed by a strong and rapid decline. However, the mechanisms responsible for this suppression are still largely unknown. Here, we show that IgG opsonization of model viruses influenza and respiratory syncytial virus (RSV) strongly and selectively suppressed type I and III IFN production by various human antigen-presenting cells. This suppression was induced by selective inhibition of TLR, RIG-I-like receptor, and STING-dependent type I and III IFN gene transcription. Surprisingly, type I and III IFN suppression was mediated by Syk and PI3K independent inhibitory signaling via FcγRIIa, thereby identifying a novel non-canonical FcγRIIa pathway in myeloid cells. Together, these results indicate that IgG opsonization of viruses functions as a novel negative feedback mechanism in humans, which may play a role in the selective suppression of type I and III IFN responses during the late-phase of viral infections. In addition, activation of this pathway may be used as a tool to limit type I IFN-associated pathology.


Subject(s)
Interferon Type I/immunology , Interferons/immunology , Myeloid Cells/immunology , Receptors, IgG/immunology , Animals , Antigen-Presenting Cells/immunology , Cells, Cultured , Female , Humans , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Respiratory Syncytial Viruses/immunology , Signal Transduction/immunology , Syk Kinase/immunology , Transcription, Genetic/immunology , Virus Diseases/immunology , Interferon Lambda
2.
Immunity ; 31(2): 181-3, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19699169

ABSTRACT

In this issue of Immunity, the studies by Sutton et al. (2009) and Martin et al. (2009) indicate that gammadelta T cells are innate cells that rapidly produce interleukin (IL)-17 in response to cytokines or pathogens without the need for T cell receptor engagement.


Subject(s)
Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cytokines/immunology , Cytokines/metabolism , Immunity, Innate , Interleukin-17/biosynthesis , Interleukin-17/immunology , Lymphocyte Activation/immunology , Mice , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/metabolism
3.
Blood ; 120(1): 112-21, 2012 Jul 05.
Article in English | MEDLINE | ID: mdl-22649103

ABSTRACT

Dendritic cells (DCs) are essential in inducing adaptive immune responses against bacteria by expressing cytokines that skew T-cell responses toward protective Th17 cells. Although it is widely recognized that induction of these cytokines by DCs involves activation of multiple receptors, it is still incompletely characterized which combination of receptors specifically skews Th17-cell responses. Here we have identified a novel role for FcγRIIa in promoting human Th17 cells. Activation of DCs by bacteria opsonized by serum IgG strongly promoted Th17 responses, which was FcγRIIa-dependent and coincided with enhanced production of selected cytokines by DCs, including Th17-promoting IL-1ß and IL-23. Notably, FcγRIIa stimulation on DCs did not induce cytokine production when stimulated individually, but selectively amplified cytokine responses through synergy with TLR2, 4, or 5. Importantly, this synergy is mediated at 2 different levels. First, TLR-FcγRIIa costimulation strongly increased transcription of pro-IL-1ß and IL-23p19. Second, FcγRIIa triggering induced activation of caspase-1, which cleaves pro-IL-1ß into its bioactive form and thereby enhanced IL-1ß secretion. Taken together, these data identified cross-talk between TLRs and FcγRIIa as a novel mechanism by which DCs promote protective effector Th17-cell responses against bacteria.


Subject(s)
Bacterial Infections/immunology , Dendritic Cells/immunology , Immunoglobulin G/immunology , Receptors, IgG/immunology , Th17 Cells/immunology , Toll-Like Receptors/immunology , Adaptive Immunity/immunology , Cell Communication/immunology , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/microbiology , Escherichia coli/immunology , Escherichia coli Infections/immunology , Humans , Ligands , Macrophages/cytology , Macrophages/immunology , Macrophages/microbiology , Receptor Cross-Talk/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Staphylococcus epidermidis/immunology , Th17 Cells/cytology , Th17 Cells/microbiology
4.
Blood ; 118(23): 6107-14, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21926348

ABSTRACT

IL-17-producing CD4(+) T helper (Th17) cells are important for immunity against extracellular pathogens and in autoimmune diseases. The factors that drive Th17 development in human remain a matter of debate. Here we show that, compared with classic CD28 costimulation, alternative costimulation via the CD5 or CD6 lymphocyte receptors forms a superior pathway for human Th17-priming. In the presence of the Th17-promoting cytokines IL-1ß, IL-6, IL-23, and transforming growth factor-ß (TGF-ß), CD5 costimulation induces more Th17 cells that produce higher amounts of IL-17, which is preceded by prolonged activation of signal transducer and activator of transcription 3 (STAT3), a key regulator in Th17 differentiation, and enhanced levels of the IL-17-associated transcription factor retinoid-related orphan receptor-γt (ROR-γt). Strikingly, these Th17-promoting signals critically depend on CD5-induced elevation of IL-23 receptor (IL-23R) expression. The present data favor the novel concept that alternative costimulation via CD5, rather than classic costimulation via CD28, primes naive T cells for stable Th17 development through promoting the expression of IL-23R.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD5 Antigens/immunology , Receptors, Interleukin/immunology , Th17 Cells/immunology , Adult , CD28 Antigens/immunology , CD28 Antigens/metabolism , CD3 Complex/immunology , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/cytology , CD5 Antigens/metabolism , Cell Differentiation/immunology , Gene Expression/immunology , Humans , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-17/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Interleukin/genetics , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Signal Transduction/immunology , Th17 Cells/cytology , Transcription, Genetic/immunology
5.
J Immunol ; 187(7): 3488-92, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21880979

ABSTRACT

The two outermost compartments of skin are populated by different Ag-presenting dendritic cell types. Epidermal Langerhans cells (LCs) are evolutionarily adapted to the continuous presence of harmless skin commensals by the selective lack of cell surface TLRs that sense bacteria. In this article, we analyze the ability of LCs and dermal dendritic cells (DDCs) to respond to virus infection. Live virus and intracellular TLR3-agonist dsRNA commit LCs more effectively than DDCs to stimulate naive CD8(+) T cell expansion and their differentiation into effector cells. This potent CD8(+) T cell-promoting capacity of LCs is causally related to high levels of virus-induced CD70 expression but not to IL-12 production. These data suggest a remarkable specialization of LCs in the induction of pathogen class-specific adaptive immunity. Whereas LCs ignore bacteria, they are superior to DDCs to initiate effective CD70-mediated CD8(+) T cells in response to virus stimulation.


Subject(s)
CD27 Ligand/immunology , CD8-Positive T-Lymphocytes/immunology , Herpesvirus 4, Human/immunology , Langerhans Cells/immunology , Lymphocyte Activation/immunology , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/virology , Cell Separation , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Dendritic Cells/virology , Flow Cytometry , Humans , Langerhans Cells/virology , Skin/cytology , Skin/immunology
6.
J Allergy Clin Immunol ; 127(6): 1532-40.e7, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21497886

ABSTRACT

BACKGROUND: The vitamin D metabolite 1,25(OH)2D3 (VitD3) is a potent immunosuppressive drug and, among others, is used for topical treatment of psoriasis. A proposed mechanism of VitD3-mediated suppression is priming of dendritic cells (DCs) to induce regulatory T (Treg) cells. OBJECTIVE: Currently, there is confusion about the phenotype of VitD3-induced Treg cells and the DC-derived molecules driving their development. We investigated Treg cell induction after VitD3 priming of 2 distinct skin DC subsets: Langerhans cells (LCs) and dermal dendritic cells (DDCs). METHODS: LCs and DDCs primed with VitD3 were cocultured with allogeneic naive T cells. The phenotype and function of the DCs and induced T cells were analyzed. RESULTS: Both VitD3-primed DC subtypes induced T cells with regulatory activity. Unexpectedly, whereas the Treg cell populations generated by VitD3-primed LCs were CD25(hi)CD127(lo) forkhead box protein 3 (Foxp3)-positive cells, which meet the criteria of classical inducible Treg cells, the T cells developing in response to VitD3-primed DDCs were Foxp3(-) T(R)1 cells expressing IL-10. Inhibition experiments revealed that LC-derived TGF-ß is a key factor in the induction of Foxp3(+) Treg cells, whereas DDC-derived IL-10 is important for the induction of IL-10(+) T(R)1 cells. CONCLUSION: Thus we report the novel finding that distinct but closely related DC subsets are differentially programmed by VitD3 to support development of either TGF-ß-dependent Foxp3(+) Treg cells or IL-10-dependent IL-10(+) Treg cells.


Subject(s)
Calcitriol/pharmacology , Langerhans Cells/drug effects , Langerhans Cells/immunology , T-Lymphocytes, Regulatory/immunology , Cell Communication , Cell Proliferation , Coculture Techniques , Cytokines/biosynthesis , Humans , Immunosuppressive Agents/pharmacology , Interleukin-10/biosynthesis , Isoantigens , Langerhans Cells/classification , Phenotype , T-Lymphocytes, Regulatory/classification , T-Lymphocytes, Regulatory/cytology , Transforming Growth Factor beta/biosynthesis
7.
J Exp Med ; 200(8): 979-90, 2004 Oct 18.
Article in English | MEDLINE | ID: mdl-15492123

ABSTRACT

The human gastric pathogen Helicobacter pylori spontaneously switches lipopolysaccharide (LPS) Lewis (Le) antigens on and off (phase-variable expression), but the biological significance of this is unclear. Here, we report that Le+ H. pylori variants are able to bind to the C-type lectin DC-SIGN and present on gastric dendritic cells (DCs), and demonstrate that this interaction blocks T helper cell (Th)1 development. In contrast, Le- variants escape binding to DCs and induce a strong Th1 cell response. In addition, in gastric biopsies challenged ex vivo with Le+ variants that bind DC-SIGN, interleukin 6 production is decreased, indicative of increased immune suppression. Our data indicate a role for LPS phase variation and Le antigen expression by H. pylori in suppressing immune responses through DC-SIGN.


Subject(s)
Cell Adhesion Molecules/physiology , Helicobacter pylori/physiology , Lectins, C-Type/physiology , Lipopolysaccharides/pharmacology , Receptors, Cell Surface/physiology , Th1 Cells/immunology , Th2 Cells/immunology , Fucosyltransferases/physiology , Humans , Interleukin-10/biosynthesis , Interleukin-6/biosynthesis , Lewis Blood Group Antigens/physiology , Lewis X Antigen/physiology , Membrane Glycoproteins/physiology , Toll-Like Receptors
8.
J Exp Med ; 195(4): 473-83, 2002 Feb 18.
Article in English | MEDLINE | ID: mdl-11854360

ABSTRACT

Dendritic cells (DCs) activated by CD40L-expressing CD4+ T cells act as mediators of "T helper (Th)" signals for CD8+ T lymphocytes, inducing their cytotoxic function and supporting their long-term activity. Here, we show that the optimal activation of DCs, their ability to produce high levels of bioactive interleukin (IL)-12p70 and to induce Th1-type CD4+ T cells, is supported by the complementary DC-activating signals from both CD4+ and CD8+ T cells. Cord blood- or peripheral blood-isolated naive CD8+ T cells do not express CD40L, but, in contrast to naive CD4+ T cells, they are efficient producers of IFN-gamma at the earliest stages of the interaction with DCs. Naive CD8+ T cells cooperate with CD40L-expressing naive CD4+ T cells in the induction of IL-12p70 in DCs, promoting the development of primary Th1-type CD4+ T cell responses. Moreover, the recognition of major histocompatibility complex class I-presented epitopes by antigen-specific CD8+ T cells results in the TNF-alpha- and IFN-gamma-dependent increase in the activation level of DCs and in the induction of type-1 polarized mature DCs capable of producing high levels of IL-12p70 upon a subsequent CD40 ligation. The ability of class I-restricted CD8+ T cells to coactivate and polarize DCs may support the induction of Th1-type responses against class I-presented epitopes of intracellular pathogens and contact allergens, and may have therapeutical implications in cancer and chronic infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Th1 Cells/immunology , CD40 Ligand/biosynthesis , CD40 Ligand/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cell Line , Dendritic Cells/cytology , Dendritic Cells/metabolism , Flow Cytometry , Histocompatibility Antigens Class I/immunology , Humans , Influenza, Human/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/pharmacology , Interleukin-12/biosynthesis , Interleukin-12/immunology , Melanoma/immunology , Th1 Cells/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
9.
J Cell Biol ; 169(4): 569-76, 2005 May 23.
Article in English | MEDLINE | ID: mdl-15897263

ABSTRACT

Langerhans cells (LC) form a unique subset of dendritic cells (DC) in the epidermis but so far their in vivo functions in skin immunity and tolerance could not be determined, in particular in relation to dermal DC (dDC). Here, we exploit a novel diphtheria toxin (DT) receptor (DTR)/DT-based system to achieve inducible ablation of LC without affecting the skin environment. Within 24 h after intra-peritoneal injection of DT into Langerin-DTR mice LC are completely depleted from the epidermis and only begin to return 4 wk later. LC deletion occurs by apoptosis in the absence of inflammation and, in particular, the dDC compartment is not affected. In LC-depleted mice contact hypersensitivity (CHS) responses are significantly decreased, although ear swelling still occurs indicating that dDC can mediate CHS when necessary. Our results establish Langerin-DTR mice as a unique tool to study LC function in the steady state and to explore their relative importance compared with dDC in orchestrating skin immunity and tolerance.


Subject(s)
Dermatitis, Contact/immunology , Diphtheria Toxin/pharmacology , Immune Tolerance/immunology , Langerhans Cells/immunology , Receptors, Cell Surface/drug effects , Skin/immunology , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Cell Death/drug effects , Cell Death/physiology , Dermatitis, Contact/physiopathology , Disease Models, Animal , Green Fluorescent Proteins , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins , Langerhans Cells/cytology , Langerhans Cells/drug effects , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/metabolism , Mice , Mice, Transgenic , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Skin/cytology
11.
J Allergy Clin Immunol ; 122(5): 969-976.e5, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18845325

ABSTRACT

BACKGROUND: Dendritic cells (DCs) can act both as innate cells in host defense and as antigen-presenting cells for naive T cells in adaptive immunity. These functions, among others, are determined by the level of production of particular cytokines. Atopic dermatitis (AD) is a chronic inflammatory skin disorder characterized by an initial phase predominated by T(H)2 cytokines that switches into a second, more chronic T(H)1-dominated eczematous phase. OBJECTIVE: To assess to what extent the AD phenotype is associated with an aberrant phenotype and function of DCs. METHODS: Classic CD1c(+)/blood DC antigen (BDCA)-1(+) myeloid (m) DCs and CD304(+)/BDCA4(+) plasmacytoid (p) DCs, the natural IFN-producing cells, were isolated from peripheral blood of patients with AD and healthy controls and analyzed for their phenotype and function. RESULTS: Purified CD1c(+)/BDCA1(+) mDCs from patients with AD showed a selective and dramatic reduction of IL-12p70 and TNF-alpha release. IL-12p70 reduction was attributed to a defective expression of both IL-12p35 and IL-12p40 subunits. Accordingly, mature CD1c(+)/BDCA1(+) mDCs from patients with AD induced considerably less IFN-gamma-producing and more IL-4-producing T(H) cells compared with mDCs from healthy controls. In addition, CD304(+)/BDCA4(+) pDCs from patients with AD produced significantly lower levels of IFN-alpha compared with healthy controls. CONCLUSION: Myeloid DCs and pDCs from patients with AD show defective IL-12, TNF-alpha, and IFN-alpha production, which may contribute to increased susceptibility to infection and to the maintenance of the T(H)2 cell-mediated allergic state in patients with AD.


Subject(s)
Dendritic Cells/immunology , Dermatitis, Atopic/immunology , Myeloid Cells/immunology , Adult , Antigens, CD1/genetics , Antigens, CD1/immunology , Antigens, Surface/genetics , Antigens, Surface/immunology , Cytokines/immunology , Dermatitis, Atopic/genetics , Female , Glycoproteins/genetics , Glycoproteins/immunology , Humans , Male , Middle Aged , Phenotype , Young Adult
12.
Front Immunol ; 10: 739, 2019.
Article in English | MEDLINE | ID: mdl-31024565

ABSTRACT

Antigen-presenting cells (APCs) such as dendritic cells (DCs) are crucial for initiation of adequate inflammatory responses, which critically depends on the cooperated engagement of different receptors. In addition to pattern recognition receptors (PRRs), Fc gamma receptors (FcγRs) have recently been identified to be important in induction of inflammation by DCs. FcγRs that recognize IgG immune complexes, which are formed upon opsonization of pathogens, induce pro-inflammatory cytokine production through cross-talk with PRRs such as Toll-like receptors (TLRs). While the physiological function of FcγR-TLR cross-talk is to provide protective immunity against invading pathogens, undesired activation of FcγR-TLR cross-talk, e.g., by autoantibodies, also plays a major role in the development of chronic inflammatory disorders such as rheumatoid arthritis (RA). Yet, the molecular mechanisms of FcγR-TLR cross-talk are still largely unknown. Here, we identified that FcγR-TLR cross-talk-induced cytokine production critically depends on activation of the transcription factor interferon regulatory factor 5 (IRF5), which results from induction of two different pathways that converge on IRF5 activation. First, TLR stimulation induced phosphorylation of TBK1/IKKε, which is required for IRF5 phosphorylation and subsequent activation. Second, FcγR stimulation induced nuclear translocation of IRF5, which is essential for gene transcription by IRF5. We identified that IRF5 activation by FcγR-TLR cross-talk amplifies pro-inflammatory cytokine production by increasing cytokine gene transcription, but also by synergistically inducing glycolytic reprogramming, which is another essential process for induction of inflammatory responses by DCs. Combined, here we identified IRF5 as a pivotal component of FcγR-TLR cross-talk in human APCs. These data may provide new potential targets to suppress chronic inflammation in autoantibody-associated diseases that are characterized by undesired or excessive FcγR-TLR cross-talk, such as RA, systemic sclerosis, and systemic lupus erythematous.


Subject(s)
Dendritic Cells/immunology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Receptors, IgG/metabolism , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Active Transport, Cell Nucleus , Dendritic Cells/metabolism , Glycolysis/immunology , Humans , I-kappa B Kinase/immunology , I-kappa B Kinase/metabolism , In Vitro Techniques , Inflammation/immunology , Macrophages/immunology , Macrophages/metabolism , Models, Immunological , Monocytes/immunology , Monocytes/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Receptor Cross-Talk/immunology , Transcription, Genetic
13.
J Immunol Methods ; 321(1-2): 94-106, 2007 Apr 10.
Article in English | MEDLINE | ID: mdl-17336322

ABSTRACT

In this study we demonstrate a novel protocol showing that electroporation of CD14+ monocytes directly isolated from blood with green fluorescent protein (GFP) RNA results in a 3-fold higher yield of antigen presenting dendritic cells (DCs) when compared to conventional methods employing immature DCs for electroporation. We further show a stable electroporation efficacy resulting in 60% of GFP positive cells. Expression of co-stimulatory molecules and maturation markers such as CD80, CD86, CD83 as well of the chemokine receptor 7 (CCR7) was found in 90% of the mature DCs. Importantly, production of IL-12p70 was 10 times higher in cells electroporated at the monocyte stage compared to cells electroporated at the immature DC stage. Stimulation of autologous naïve lymphocytes by DCs electroporated at monocytes stage elicited proliferation of CD8+ T-cell with 7-fold increase in IFN-gamma release. Blocking of the MHC-Class I molecules significantly inhibited the IFN-gamma release, indicating that antigen presentation was MHC-Class I mediated. In summary, electroporation of CD14+ monocytes with RNA results in a high yield of antigen presenting DCs with high immuno-stimulatory capacity and antigen presentation on MHC-Class I molecules. This improved method may represent an attractive approach for RNA-based DC immunotherapy.


Subject(s)
Dendritic Cells/immunology , Electroporation , Lipopolysaccharide Receptors/metabolism , Monocytes/immunology , Transfection/methods , Adult , Antigens, CD/metabolism , CD4-CD8 Ratio , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Communication/immunology , Cell Differentiation , Cell Survival , Cells, Cultured , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/metabolism , Female , Genes, Reporter , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Histocompatibility Antigens Class I/immunology , Humans , Interferon-gamma/biosynthesis , Interleukin-10/metabolism , Interleukin-12/biosynthesis , Lymphocyte Activation , Male , Monocytes/cytology , Monocytes/metabolism , RNA/genetics , RNA/metabolism , Receptors, CCR7 , Receptors, Chemokine/biosynthesis , Time Factors
14.
Curr Opin Allergy Clin Immunol ; 7(5): 374-81, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17873575

ABSTRACT

PURPOSE OF REVIEW: Recently, a novel and unique subset of interleukin (IL)-17-producing CD4+ T helper (Th17) cells, distinct from Th1 and Th2 cells, was discovered. The question is addressed as to what extent inflammatory skin diseases are associated with the actions of this newly discovered Th17 cell subset. RECENT FINDINGS: Th17 cells are involved in protection against bacterial pathogens. In addition, it is now clear that Th17 cells may also be crucial in the pathogenesis of various chronic inflammatory diseases that were formerly categorized as Th1-mediated disorders. SUMMARY: In this review, we summarize the current knowledge of IL-17 and Th17 cells and discuss the possible role of IL-17 in the pathology of psoriasis, contact hypersensitivity and atopic dermatitis. Whereas IL-17 may play an important role in the pathogenesis of psoriasis and contact hypersensitivity, its role in atopic dermatitis is still unclear.


Subject(s)
Dermatitis, Atopic/immunology , Dermatitis, Contact/immunology , Interleukin-17/immunology , Psoriasis/immunology , T-Lymphocyte Subsets/immunology , Animals , Dermatitis, Atopic/metabolism , Dermatitis, Contact/metabolism , Humans , Immunity, Innate , Interleukin-17/metabolism , Psoriasis/metabolism , Skin/immunology , T-Lymphocyte Subsets/metabolism
15.
Cancer Res ; 64(17): 5934-7, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15342370

ABSTRACT

Using the principle of functional polarization of dendritic cells (DCs), we have developed a novel protocol to generate human DCs combining the three features critical for the induction of type-1 immunity: (a) fully mature status; (b) responsiveness to secondary lymphoid organ chemokines; and (c) high interleukin-12p70 (IL-12p70)-producing ability. We show that IFN-alpha and polyinosinic:polycytidylic acid (p-I:C) synergize with the "classical" type-1-polarizing cytokine cocktail [tumor necrosis factor alpha (TNFalpha)/IL-1beta/IFNgamma], allowing for serum-free generation of fully mature type-1-polarized DCs (DC1). Such "alpha-type-1-polarized DC(s)" (alphaDC1) show high migratory responses to the CCR7 ligand, 6C-kine but produce much higher levels of IL-12p70 as compared to TNFalpha/IL-1beta/IL-6/prostaglandin E2 (PGE2)-matured DCs (sDC), the current "gold standard" in DC-based cancer vaccination. A single round of in vitro sensitization with alphaDC1 (versus sDCs) induces up to 40-fold higher numbers of long-lived CTLs against melanoma-associated antigens: MART-1, gp100, and tyrosinase. Serum-free generation of alphaDC1 allows, for the first time, the clinical application of DCs that combine the key three features important for their efficacy as anticancer vaccines.


Subject(s)
Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , T-Lymphocytes, Cytotoxic/immunology , Cell Polarity/immunology , Chemotaxis/immunology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Humans , Interferon-alpha/immunology , Interferon-alpha/pharmacology , Interferon-gamma/immunology , Interferon-gamma/pharmacology , Interleukin-1/immunology , Interleukin-1/pharmacology , Interleukin-12/biosynthesis , Interleukin-12/immunology , Ligands , Melanoma/blood , Melanoma/immunology , Poly I-C/immunology , Poly I-C/pharmacology , Protein Subunits/biosynthesis , Protein Subunits/immunology , Receptors, CCR7 , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology
16.
J Mol Med (Berl) ; 82(3): 197-205, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14673529

ABSTRACT

The reaction of the intestinal immune system to intestinal bacteria shows striking differences between various bacterial strains. Whereas Klebsiella pneumoniae induces a fierce proinflammatory reaction, the probiotic strain Lactobacillus rhamnosus has clear anti-inflammatory effect in gastrointestinal disease and allergy. The molecular basis for this dichotomy is poorly understood but is likely to involve different modulation of antigen-presenting dendritic cells (DC) by L. rhamnosus and K. pneumoniae. Hence we evaluated phenotypic and functional characteristics of DC matured in the presence of L. rhamnosus and K. pneumoniae. Monocyte-derived immature DC were cultured in the presence of live bacteria to obtain mature DC. Both micro-organisms induced maturation of immature DC as shown by CD83 and CD86 expression, but receptors involved in activation of Th1 cells were expressed predominantly on DC exposed to K. pneumoniae. In contrast to K. pneumoniae, maturation with L. rhamnosus resulted in lower TNF-alpha, IL-6, and IL-8 production by immature DC and lower IL-12 and IL-18 production by mature DC. Moreover, L. rhamnosus led to the development of T cells without a typical Th phenotype whereas K. pneumoniae induced a Th1 immune response, dependent mainly on IL-12 production. Thus our results strongly support the concept that differential modulation of DC explains the differences in the immune response to various bacterial strains and indicates that K. pneumoniae induces Th1 immune responses via DC.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/microbiology , Klebsiella pneumoniae/metabolism , Lactobacillus/metabolism , Antigens, CD/biosynthesis , Antigens, CD1/biosynthesis , B7-2 Antigen , Cell Separation , Cytokines/biosynthesis , Dendritic Cells/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Immunoglobulins/biosynthesis , Inflammation , Interleukin-12/biosynthesis , Interleukin-18/biosynthesis , Lipopolysaccharide Receptors/biosynthesis , Membrane Glycoproteins/biosynthesis , Monocytes/cytology , Monocytes/microbiology , Phenotype , Protein Subunits/biosynthesis , T-Lymphocytes/metabolism , CD83 Antigen
17.
Immunobiology ; 220(2): 193-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25108563

ABSTRACT

Myeloid antigen-presenting cells (APCs) tailor immune responses to the pathogen involved through the production of specific pro- and anti-inflammatory cytokines. It is becoming increasingly clear that the ultimate cytokine profile produced by myeloid APCs crucially depends on interaction between multiple pathogen recognizing receptors. In this respect, we recently identified an important role for cross-talk between Fc gamma receptor IIa (FcγRIIa) and Toll-like receptors (TLRs) in human dendritic cells (DCs), which induces anti-bacterial immunity through the selective induction of TNFα and Th17-promoting cytokines. Here, we show that FcγRIIa-TLR cross-talk is not restricted to DCs, but is a common feature of various human myeloid APC subsets including monocytes and macrophages. Interestingly, FcγRIIa-TLR cross-talk in monocytes resulted in the induction of a cytokine profile distinct from that in DCs and macrophages, indicating that FcγRIIa stimulation induces cell-type and tissue specific responses. Surprisingly, we show that the FCGR2A H131R single nucleotide polymorphism (SNP), which is known to greatly affect FcγRIIa-mediated uptake of IgG2-opsonized bacteria, did not affect FcγRIIa-dependent cytokine production, indicating that these processes are differently regulated. In addition, we demonstrate that FcγRIIa selectively synergized with TLRs, IL-1R, and IFNγR, but did not affect cytokine production induced by other receptors such as C-type lectin receptor Dectin-1. Taken together, these data demonstrate that FcγRIIa-dependent modulation of cytokine production is more widespread than previously considered, and indicate that cross-talk of FcγRIIa with various receptors and in multiple cell types contributes to the induction of pathogen and tissue-specific immunity.


Subject(s)
Cytokines/biosynthesis , Myeloid Cells/immunology , Myeloid Cells/metabolism , Receptors, IgG/metabolism , Receptors, Interferon/metabolism , Receptors, Interleukin-1/metabolism , Toll-Like Receptors/metabolism , Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/metabolism , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunomodulation , Leukocytes, Mononuclear , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Organ Specificity , Polymorphism, Single Nucleotide , Receptor Cross-Talk , Receptors, IgG/genetics , Signal Transduction
18.
J Invest Dermatol ; 120(6): 990-7, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12787126

ABSTRACT

Dendritic cells play a key role in establishing the class of immune response against invading pathogens. Upon engagement with double-stranded RNA, a major bioactive constituent of many virus types, immature dendritic cells develop into type 1 immunostimulatory dendritic cells that promote Th1 responses. Immature dendritic cells reside in the epithelia and are in close contact with keratinocytes. We studied to what extent dendritic cells can also adopt a type 1 immunostimulatory dendritic cell phenotype indirectly, as a result of the interaction with keratinocytes responding to double-stranded RNA. In contrast to supernatants from keratinocytes activated by the combination of tumor necrosis factor alpha and interleukin-1beta, supernatants from keratinocytes activated by synthetic double-stranded RNA, polyriboinosinic polyribocytidylic acid, comprised tumor necrosis factor alpha and type I interferons, which induced maturation of human monocyte-derived immature dendritic cells. In addition, dendritic cells matured in the presence of these supernatants strongly biased the development of Th1 cells from naive Th cells. This bias was dependent on keratinocyte-derived interferon-alpha/beta and interleukin-18, as neutralization of both interferon-alpha/beta and interleukin-18 in the keratinocyte culture supernatant reduced the development of interferon-gamma-producing Th cells. These findings suggest that keratinocytes can contribute to the development of selective Th1/Th2 responses through the induction of maturation and functional polarization of dendritic cells, indicating a novel role for keratinocytes as initiators and regulators of cutaneous T-cell-mediated inflammation. In addition, these results support the concept that, in addition to direct interaction with pathogens, dendritic cells may also be activated and primed by pathogen indirectly, via the effect of resident tissue cells responding to pathogen.


Subject(s)
Dendritic Cells/physiology , Keratinocytes/drug effects , Keratinocytes/physiology , RNA, Double-Stranded/pharmacology , Th1 Cells/physiology , Cell Division/physiology , Cell Polarity , Cells, Cultured , Cellular Senescence/physiology , Humans , Interferon Type I/physiology , Interleukin-1/pharmacology , Interleukin-12/physiology , Interleukin-18/physiology , Phenotype , Poly I-C/pharmacology , Th1 Cells/cytology , Th1 Cells/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/physiology
19.
Vaccine ; 32(47): 6294-302, 2014 Oct 29.
Article in English | MEDLINE | ID: mdl-25236584

ABSTRACT

Vitamin D is recognized as a potent immunosuppressive drug. The suppressive effects of vitamin D are attributed to its physiologically active metabolite 1,25 dihydroxy vitamin D3 (calcitriol), which was shown, to prime dendritic cells (DCs) to promote the development of regulatory T (Treg) cells. Despite the potential benefit in treating autoimmune diseases, clinical application of calcitriol is hindered by deleterious side effects manifested by hypercalcemia and hypercalciuria. Conversely, the physiological precursors of calcitriol, vitamin D3 (cholecalciferol) and its first metabolite 25-hydroxy vitamin D3 (calcidiol) are widely applied in the clinic due to their low calcimic burden. However, the mechanisms by which cholecalciferol and calcidiol may modulate adaptive immunity remain elusive. This prompted us to unravel the immunosuppressive capacity of these precursors by assessing their influence on DC functions and the subsequent polarization of naïve CD4(+) T cells. In this study we show that, whereas cholecalciferol has insignificant effects on DC maturation and cytokine production, it only weakly primed DCs to induce suppressive T cells. However, like calcitriol, calcidiol not only exerted an inhibitory effect on DC maturation and cytokine production, and primed DCs to promote the development of suppressive IL-10-producing Treg cells. Strikingly, in contrast to the population of IL-10-producing Treg cells induced by calcitriol-primed DCs, the IL-10-producing Treg cells induced by calcidiol-primed DCs exhibited sustained IFN-γ production in face of their suppressive capacity. Experiments with the steroid synthesis inhibitor ketoconazole indicated that the immunomodulatory features of the precursors are dependent on their conversion into calcitriol. Collectively, calcidiol is a potent immune modulator, which may be more adequate than calcitriol for the treatment of chronic inflammatory diseases, since it is less hypercalcimic. This may be of particular interest for the treatment of allergic disease, where concurrent suppression and sustained IFN-γ production by Treg cells effectively counterbalance the Th2-dominated immune responses.


Subject(s)
Calcifediol/pharmacology , Dendritic Cells/drug effects , Interleukin-10/biosynthesis , T-Lymphocytes, Regulatory/immunology , Cells, Cultured , Cholecalciferol/pharmacology , Coculture Techniques , Dendritic Cells/immunology , Humans , Interferon-gamma/biosynthesis
20.
Nat Commun ; 5: 5444, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25392121

ABSTRACT

M2 macrophages suppress inflammation in numerous disorders, including tumour formation, infection and obesity. However, the exact role of M2 macrophages in the context of several other diseases is still largely undefined. We here show that human M2 macrophages promote inflammation instead of suppressing inflammation on simultaneous exposure to complexed IgG (c-IgG) and TLR ligands, as occurs in the context of diseases such as rheumatoid arthritis (RA). c-IgG-TLR ligand co-stimulation of M2 macrophages selectively amplifies production of pro-inflammatory cytokines TNF-α, IL-1ß and IL-6 and promotes Th17 responses, which all play a critical role in RA pathology. Induction of pro-inflammatory cytokines on c-IgG co-stimulation mainly depends on Fc gamma receptor IIa (FcγRIIa), which selectively amplifies cytokine gene transcription and induces caspase-1 activation. These data indicate that FcγR-TLR cross-talk may be targeted for treatment to attenuate inflammation in RA, by restoring the anti-inflammatory function of M2 macrophages.


Subject(s)
Inflammation/physiopathology , Interleukin-1beta/physiology , Interleukin-6/physiology , Macrophages/physiology , Receptor Cross-Talk/physiology , Receptors, IgG/physiology , Tumor Necrosis Factor-alpha/physiology , Caspase 1/metabolism , Enzyme Activation/physiology , Gene Expression Regulation/physiology , Humans , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Macrophages/metabolism , Receptors, IgG/metabolism , Th17 Cells/physiology , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL