Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
Front Immunol ; 9: 869, 2018.
Article in English | MEDLINE | ID: mdl-29760700

ABSTRACT

Avian influenza A of the subtype H7N9 has been responsible for almost 1,600 confirmed human infections and more than 600 deaths since its first outbreak in 2013. Although sustained human-to-human transmission has not been reported yet, further adaptations to humans in the viral genome could potentially lead to an influenza pandemic, which may have severe consequences due to the absence of pre-existent immunity to this strain at population level. Currently there is no influenza A (H7N9) vaccine available. Therefore, in case of a pandemic outbreak, alternative preventive approaches are needed, ideally even independent of the type of influenza virus outbreak. Bacillus Calmette-Guérin (BCG) is known to induce strong heterologous immunological effects, and it has been shown that BCG protects against non-related infection challenges in several mouse models. BCG immunization of mice as well as human induces trained innate immune responses, resulting in increased cytokine responses upon subsequent ex vivo peripheral blood mononuclear cell restimulation. We investigated whether BCG (Statens Serum Institut-Denmark)-induced trained immunity may protect against a lethal avian influenza A/Anhui/1/2013 (H7N9) challenge. Here, we show that isolated splenocytes as well as peritoneal macrophages of BCG-immunized BALB/c mice displayed a trained immunity phenotype resulting in increased innate cytokine responses upon ex vivo restimulation. However, after H7N9 infection, no significant differences were found between the BCG immunized and the vehicle control group at the level of survival, weight loss, pulmonary influenza A nucleoprotein staining, or histopathology. In conclusion, BCG-induced trained immunity did not result in protection in an oseltamivir-sensitive influenza A/Anhui/1/2013 (H7N9) challenge mouse model.


Subject(s)
BCG Vaccine/immunology , Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/immunology , Animals , Female , Mice , Mice, Inbred BALB C , Vaccination
3.
Proc Natl Acad Sci U S A ; 114(8): E1480-E1489, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28167776

ABSTRACT

TGF-ß is an anti-inflammatory cytokine whose signaling is negatively controlled by Smad7. Previously, we established a role for Smad7 in the generation of autoreactive T cells; however, the function of Smad7 in dendritic cells (DCs) remains elusive. Here, we demonstrate that DC-specific Smad7 deficiency resulted in elevated expression of the transcription factors Batf3 and IRF8, leading to increased frequencies of CD8+CD103+ DCs in the spleen. Furthermore, Smad7-deficient DCs expressed higher levels of indoleamine 2,3-dioxygenase (IDO), an enzyme associated with tolerance induction. Mice devoid of Smad7 specifically in DCs are resistant to the development of experimental autoimmune encephalomyelitis (EAE) as a result of an increase of protective regulatory T cells (Tregs) and reduction of encephalitogenic effector T cells in the central nervous system. In agreement, inhibition of IDO activity or depletion of Tregs restored disease susceptibility. Intriguingly, when Smad7-deficient DCs also lacked the IFN-γ receptor, the mice regained susceptibility to EAE, demonstrating that IFN-γ signaling in DCs mediates their tolerogenic function. Our data indicate that Smad7 expression governs splenic DC subset differentiation and is critical for the promotion of their efficient function in immunity.


Subject(s)
Autoimmunity/physiology , Dendritic Cells/metabolism , Smad7 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/physiology , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Immune Tolerance , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon Regulatory Factors/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/physiology , Spleen/metabolism , T-Lymphocytes, Regulatory/metabolism
4.
Oncotarget ; 7(22): 32015-30, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27027442

ABSTRACT

Although IL-10 promotes a regulatory phenotype of CD11c+ dendritic cells and macrophages in vitro, the role of IL-10 signaling in CD11c+ cells to maintain intestinal tolerance in vivo remains elusive. To this aim, we generated mice with a CD11c-specific deletion of the IL-10 receptor alpha (Cd11ccreIl10rafl/fl). In contrast to the colon, the small intestine of Cd11ccreIl10rafl/fl mice exhibited spontaneous crypt hyperplasia, increased numbers of intraepithelial lymphocytes and lamina propria T cells, associated with elevated levels of T cell-derived IFNγ and IL-17A. Whereas naive mucosal T-cell priming was not affected and oral tolerance to ovalbumin was intact, augmented T-cell function in the lamina propria was associated with elevated numbers of locally dividing T cells, expression of T-cell attracting chemokines and reduced T-cell apoptosis. Upon stimulation, intestinal IL-10Rα deficient CD11c+ cells exhibited increased activation associated with enhanced IL-6 and TNFα production. Following colonization with Helicobacter hepaticus Cd11ccreIl10rafl/fl mice developed severe large intestinal inflammation characterized by infiltrating T cells and increased levels of Il17a, Ifng, and Il12p40. Altogether these findings demonstrate a critical role of IL-10 signaling in CD11c+ cells to control small intestinal immune homeostasis by limiting reactivation of local memory T cells and to protect against Helicobacter hepaticus-induced colitis.


Subject(s)
CD11c Antigen/metabolism , Colitis/prevention & control , Helicobacter Infections/prevention & control , Immunity, Mucosal , Interleukin-10/metabolism , Intestinal Mucosa/metabolism , Intestine, Large/metabolism , Intestine, Small/metabolism , T-Lymphocytes/metabolism , Animals , CD11c Antigen/deficiency , CD11c Antigen/genetics , CD11c Antigen/immunology , Colitis/immunology , Colitis/metabolism , Colitis/microbiology , Disease Models, Animal , Genetic Predisposition to Disease , Helicobacter Infections/immunology , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter hepaticus/immunology , Helicobacter hepaticus/pathogenicity , Homeostasis , Immunologic Memory , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-10/immunology , Interleukin-10 Receptor alpha Subunit/genetics , Interleukin-10 Receptor alpha Subunit/immunology , Interleukin-10 Receptor alpha Subunit/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestine, Large/immunology , Intestine, Large/microbiology , Intestine, Small/immunology , Intestine, Small/microbiology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/microbiology
5.
J Allergy Clin Immunol ; 129(1): 143-50.e1-10, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21975175

ABSTRACT

BACKGROUND: IL-10 is a pleiotropic cytokine and potent negative regulator of both innate and adaptive immune responses. Consequently, IL-10-deficient (IL-10(-/-)) mice have enhanced contact hypersensitivity (CHS) to topical hapten. OBJECTIVE: Although the importance of IL-10 production by (regulatory) T cells and Langerhans cells in regulating CHS has been established by cell type-specific il-10 gene targeting, it remains elusive to what extent IL-10 controls dendritic cell (DC) function in vivo. METHODS: To this aim, we generated mice with a DC-specific deletion of the IL-10 receptor (IL-10R). RESULTS: Despite the ability of IL-10 to inhibit DC maturation in vitro, DCs of resting DC-IL10R(-/-) mice retained their immature phenotype in vivo. In contrast, IL-10R(-/-) DCs produced increased levels of proinflammatory cytokines and IL-10 after in vitro stimulation. Induction of CHS was indistinguishable from that seen in control mice at 24 hours after hapten challenge but resulted in increased ear swelling at 48 hours and delayed resolution of the inflammatory reaction. Adoptive T-cell transfer experiments revealed that only T-cell reactivation and not sensitization by IL-10R(-/-) DCs leads to enhanced CHS. Accordingly, the expression of proinflammatory cytokines and IL-10 was augmented in the skin of DC-IL10R(-/-) mice after hapten challenge. CONCLUSION: Our data demonstrate that IL-10 signaling in DCs is dispensable during naive T-cell priming but is critical to prevent an exaggerated effector T-cell response in the skin.


Subject(s)
Dendritic Cells/immunology , Dermatitis, Contact/immunology , Interleukin-10/physiology , Lymphocyte Activation/immunology , Skin/immunology , T-Lymphocytes/immunology , Animals , Cytokines/biosynthesis , Dendritic Cells/metabolism , Dermatitis, Contact/genetics , Gene Deletion , Immunity, Innate , Mice , Mice, Knockout , Phenotype , Receptors, Interleukin-10/genetics , Signal Transduction , Skin/metabolism
6.
J Immunol ; 187(10): 5069-76, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21998450

ABSTRACT

The critical role of Langerhans cells (LC) in contact hypersensitivity (CHS) was recently questioned in studies using different LC-depletion mouse models. On one hand, inducible ablation of LC led to diminished ear swelling, suggesting functional redundancy between LC and (Langerin(+)) dermal dendritic cells (DC). On the other hand, constitutive or acute depletion of LC resulted in an enhanced reaction, supporting a regulatory role of LC in CHS. To address this controversy by conditional gene targeting, we generated Langerin-Cre knockin mice. Breeding these mice to a Cre-reporter strain demonstrated robust and specific DNA recombination in LC, as well as other Langerin(+) tissue DC. In agreement with the vital requirement of TGF-ß signaling for LC development, crossing Langerin-Cre to mice homozygous for a loxP-flanked TGF-ßR1 allele resulted in permanent LC deficiency, whereas the homeostasis of dermal Langerin(+) DC was unaffected. In the absence of LC, induction of CHS in these Langerin(+) DC-specific TGF-ßR1-deficient mice elicited decreased ear swelling compared with controls. This novel approach provided further evidence against a regulatory function of LC in CHS. Moreover, these Langerin-Cre mice represent a unique and powerful tool to dissect the role and molecular control of Langerin(+) DC populations beyond LC.


Subject(s)
Antigens, CD/genetics , Dermatitis, Allergic Contact/immunology , Gene Deletion , Integrases/genetics , Langerhans Cells/immunology , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Mannose-Binding Lectins/deficiency , Mannose-Binding Lectins/genetics , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , Animals , Antigens, CD/biosynthesis , Dendritic Cells/enzymology , Dendritic Cells/immunology , Dendritic Cells/pathology , Dermatitis, Allergic Contact/genetics , Dermatitis, Allergic Contact/pathology , Disease Models, Animal , Gene Expression Regulation/immunology , Gene Knock-In Techniques/methods , Gene Targeting , Langerhans Cells/enzymology , Langerhans Cells/pathology , Lectins, C-Type/biosynthesis , Mannose-Binding Lectins/biosynthesis , Mice , Mice, Knockout , Mice, Transgenic , Protein Serine-Threonine Kinases/physiology , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/physiology
7.
J Immunol ; 185(6): 3248-55, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20713882

ABSTRACT

The pivotal role of TGF-beta in Langerhans cell (LC) development has been previously established in TGF-beta-deficient mice, which lack epidermal LCs. As to whether TGF-beta also governs LC homeostasis and function remains elusive. To assess the role of TGF-beta-mediated control of cutaneous dendritic cells (DCs) in vivo, we generated mice with a conditional knockout of the TGF-beta receptor 1 (TbetaR1) under a DC-specific promoter (DC-TbetaR1(del) mice). While initial LC seeding occurred in DC-TbetaR1(del) mice, the cells disappeared from the epidermis during the first week of life. TbetaR1-deficient LCs demonstrated spontaneous maturation and gained migratory potential based on increased surface expression of MHC class II, costimulatory molecules, and CCR7 and downregulation of E-cadherin. In parallel to their early loss from the epidermis, migrating LCs were reduced in the dermis and skin-draining lymph nodes of adult DC-TbetaR1(del) mice, whereas the number of Langerin(+) dermal DCs was similar to wild-type. In the absence of LCs, low-dose contact hypersensitivity in DC-TbetaR1(del) mice was significantly diminished. In contrast, ear swelling was restored to wild-type levels when a higher hapten dose was applied to efficiently target TbetaR1-deficient dermal DCs. In conclusion, TGF-beta inhibits in vivo LC maturation and migratory phenotype, identifying TGF-beta as a critical factor controlling LC homeostasis in the steady state.


Subject(s)
Cell Differentiation/immunology , Epidermal Cells , Epidermis/immunology , Langerhans Cells/cytology , Langerhans Cells/immunology , Transforming Growth Factor beta1/physiology , Animals , Cell Differentiation/genetics , Cell Movement/genetics , Cell Movement/immunology , Cells, Cultured , Dermatitis, Contact/immunology , Dermatitis, Contact/metabolism , Dermatitis, Contact/pathology , Epidermis/pathology , Homeostasis/genetics , Homeostasis/immunology , Langerhans Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics
8.
Immunol Cell Biol ; 88(4): 351-60, 2010.
Article in English | MEDLINE | ID: mdl-20351747

ABSTRACT

Langerhans cells (LC) are members of the heterogenous family of professional antigen presenting dendritic cells (DC). They are identified by the C-type lectin receptor Langerin and form a contiguous network in the epidermis. Consequently, LC are an integral part of the skin barrier to the environment and were considered to be critical inducers of skin immunity, whereas dermal DC were largely overlooked. However, with the identification of a distinct subset of Langerin expressing dermal DC, the situation in the skin has become more complex and the relative contribution of the different cutaneous DC populations in balancing immunity and tolerance has become a matter of active debate. Here, we briefly review the classical paradigm and recent challenges of LC function, before focusing on advances concerning their role in contact hypersensitivity and ultraviolet radiation-induced immunosuppression obtained with in vivo LC ablation models. We then discuss novel LC/DC-specific gene targeting approaches currently used to dissect the role of the regulatory cytokines transforming growth factor-beta and interleukin-10 to govern LC and DC function in vivo. This second generation of LC-specific genetically engineered mice will considerably extend our understanding of the molecular control of LC function in regulating skin immunity and tolerance in the near future.


Subject(s)
Langerhans Cells/immunology , Skin/immunology , Animals , Immunity
9.
J Leukoc Biol ; 84(1): 182-90, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18424662

ABSTRACT

Tolerance to experimental autoimmune encephalomyelitis (EAE) in SJL mice can be induced by immunization with a mannosylated form of the proteolipid protein (M-PLP139-151), despite the presence of CFA. The state of tolerance is characterized by poor delayed-type hypersensitivity responses and the absence of clinical EAE symptoms. In vivo monitoring of CFSE-labeled PLP139-151-specific TCR-transgenic (5B6) T cells revealed that immunization with M-PLP139-151 increases the clonal expansion of 5B6 T cells that do not develop full effector functions. Moreover, nonfunctional T cells obtained from M-PLP139-151-immunized mice showed poor blastogenesis and were unable to transfer EAE to naïve recipients. Nevertheless, the in vitro production of cytokines and chemokines associated with EAE was unaffected. Importantly, tolerance induced by M-PLP139-151 was abrogated by the administration of pertussis toxin, resulting in EAE development. Our results suggest that M-PLP139-151 inhibits EAE development by affecting the differentiation of T cells into encephalitogenic effector cells.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/prevention & control , Mannose/metabolism , Peptides/immunology , T-Lymphocytes/cytology , Animals , Cell Count , Cell Proliferation/drug effects , Clone Cells , Encephalomyelitis, Autoimmune, Experimental/immunology , Epitopes , Female , Immune Tolerance , Immunization , Mice , Pertussis Toxin/pharmacology , Spleen/drug effects , Spleen/pathology , T-Lymphocytes/drug effects
10.
Int Immunol ; 20(1): 117-27, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18024466

ABSTRACT

In this study, we investigated the development of T cell responses in mice after administration of a mannosylated ovalbumin peptide (M-OVA(323-339)). Immunization with M-OVA(323-339) in complete adjuvant resulted in enhanced antigen presentation in draining lymph nodes. Monitoring the fate of CFSE-labeled ovalbumin peptide-specific TCR transgenic CD4(+) T cells revealed that immunization with M-OVA(323-339) induced normal clonal expansion, recirculation and CD62L expression of antigen-specific T cells in vivo. However, these T cells developed only poor effector functions, reflected by minimal IFN-gamma production, low IgG2a levels in serum and poor peptide-specific delayed-type hypersensitivity (DTH) responses. This diminished inflammatory response was associated with decreased infiltration of T cell blasts and macrophages. Importantly, also mice with functional effector T cells did not mount a robust DTH response after a challenge with M-OVA(323-339) in the ear, although their T cells responded normally to M-OVA(323-339) in vitro. In conclusion, mannosylated peptide induces proliferation of T cells with impaired T(h)1 cell effector functions and additionally abrogates the activity of pre-existing effector T cells.


Subject(s)
Antigen Presentation , Hypersensitivity, Delayed , Mannose , Ovalbumin , Peptides , Th1 Cells/immunology , Amino Acid Sequence , Animals , Female , Immunization , Lymphocyte Activation , Mannose/administration & dosage , Mannose/chemical synthesis , Mannose/chemistry , Mannose/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Ovalbumin/administration & dosage , Ovalbumin/chemical synthesis , Ovalbumin/chemistry , Ovalbumin/immunology , Peptides/administration & dosage , Peptides/chemical synthesis , Peptides/chemistry , Peptides/immunology , Receptors, Antigen, T-Cell/genetics
11.
J Neuroimmunol ; 160(1-2): 178-87, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15710471

ABSTRACT

SJL mice immunized with mannosylated (M-) PLP(139-151) in complete adjuvant do not develop EAE and little CNS mononuclear cell infiltration; other mannosylated peptides were ineffective in this experimental setting. Despite apparently normal T cell responses, M-PLP(139-151)-immunized mice show impaired delayed-type-sensitivity to PLP(139-151) but a normal response to other peptides. After re-immunization with PLP(139-151) in complete adjuvant, these mice are largely tolerant to EAE, show less T cell proliferation and decreased peptide-specific IgG2a. Our data suggest that M-PLP(139-151) induces peptide-specific tolerance to EAE via a mechanism of deletion or impaired migration of encephalitogenic T cells.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Mannose/metabolism , Myelin Proteolipid Protein/administration & dosage , Myelin Proteolipid Protein/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Self Tolerance/immunology , Amino Acid Sequence , Animals , Cell Movement/immunology , Cells, Cultured , Central Nervous System/immunology , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/pathology , Immunodominant Epitopes/immunology , Injections, Subcutaneous , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Mice , Molecular Sequence Data , Myelin Proteolipid Protein/metabolism , Peptide Fragments/metabolism , Severity of Illness Index , Th1 Cells/immunology , Th1 Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...