Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Publication year range
1.
Eur J Immunol ; 53(11): e2249816, 2023 11.
Article in English | MEDLINE | ID: mdl-36303448

ABSTRACT

This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy, and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs and various non-lymphoid tissues. This article provides protocols with top ticks and pitfalls for preparation and successful generation of mouse and human DC from different cellular sources, such as murine BM and HoxB8 cells, as well as human CD34+ cells from cord blood, BM, and peripheral blood or peripheral blood monocytes. We describe murine cDC1, cDC2, and pDC generation with Flt3L and the generation of BM-derived DC with GM-CSF. Protocols for human DC generation focus on CD34+ cell culture on OP9 cell layers for cDC1, cDC2, cDC3, and pDC subset generation and DC generation from peripheral blood monocytes (MoDC). Additional protocols include enrichment of murine DC subsets, CRISPR/Cas9 editing, and clinical grade human DC generation. While all protocols were written by experienced scientists who routinely use them in their work, this article was also peer-reviewed by leading experts and approved by all co-authors, making it an essential resource for basic and clinical DC immunologists.


Subject(s)
Dendritic Cells , Monocytes , Animals , Mice , Humans , Antigens, CD34 , Phenotype , Cell Differentiation
2.
Eur J Immunol ; 52(11): 1712-1749, 2022 11.
Article in English | MEDLINE | ID: mdl-35099816

ABSTRACT

DCs do not just excel in antigen presentation. They orchestrate information transfer from innate to adaptive immunity by sensing and integrating a variety of danger signals, and translating them to naïve T cells, to mount specifically tailored immune responses. This is accomplished by distinct DC types specialized in different functions and because each DC is functionally plastic, assuming different activation states depending on the input signals received. Mouse models hold the key to untangle this complexity and determine which DC types and activation states contribute to which functions. Here, we aim to provide comprehensive information for selecting the most appropriate mutant mouse strains to address specific research questions on DCs, considering three in vivo experimental approaches: (i) interrogating the roles of DC types through their depletion; (ii) determining the underlying mechanisms by specific genetic manipulations; (iii) deciphering the spatiotemporal dynamics of DC responses. We summarize the advantages, caveats, suggested use, and perspectives for a variety of mutant mouse strains, discussing in more detail the most widely used or accurate models. Finally, we discuss innovative strategies to improve targeting specificity and next-generation mutant mouse models, and briefly address how humanized mouse models can accelerate translation into the clinic.


Subject(s)
Antigen Presentation , Dendritic Cells , Mice , Animals , T-Lymphocytes , Adaptive Immunity , Disease Models, Animal
3.
Cell ; 135(1): 37-48, 2008 Oct 03.
Article in English | MEDLINE | ID: mdl-18854153

ABSTRACT

Plasmacytoid dendritic cells (PDCs) represent a unique immune cell type specialized in type I interferon (IFN) secretion in response to viral nucleic acids. The molecular control of PDC lineage specification has been poorly understood. We report that basic helix-loop-helix transcription factor (E protein) E2-2/Tcf4 is preferentially expressed in murine and human PDCs. Constitutive or inducible deletion of murine E2-2 blocked the development of PDCs but not of other lineages and abolished IFN response to unmethylated DNA. Moreover, E2-2 haploinsufficiency in mice and in human Pitt-Hopkins syndrome patients was associated with aberrant expression profile and impaired IFN response of the PDC. E2-2 directly activated multiple PDC-enriched genes, including transcription factors involved in PDC development (SpiB, Irf8) and function (Irf7). These results identify E2-2 as a specific transcriptional regulator of the PDC lineage in mice and humans and reveal a key function of E proteins in the innate immune system.


Subject(s)
Dendritic Cells/immunology , Nerve Tissue Proteins/immunology , TCF Transcription Factors/immunology , Adolescent , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Child , Child, Preschool , DNA-Binding Proteins , Dendritic Cells/metabolism , Humans , Hyperventilation/immunology , Immunity, Innate , Intellectual Disability/immunology , Interferons/immunology , Mice , Syndrome , Transcription Factor 4 , Transcription Factor 7-Like 2 Protein , Transcription Factors
4.
Int J Mol Sci ; 21(2)2020 Jan 11.
Article in English | MEDLINE | ID: mdl-31940843

ABSTRACT

Contact hypersensitivity (CHS) is an established animal model for allergic contact dermatitis. Dendritic cells (DCs) play an important role in the sensitization phase of CHS by initiating T cell responses to topically applied haptens. The cannabinoid receptors 1 (CB1) and 2 (CB2) modulate DC functions and inflammatory skin responses, but their influence on the capacity of haptenized DCs to induce CHS is still unknown. We found lower CHS responses to 2,4-dinitro-1-fluorobenzene (DNFB) in wild type (WT) mice after adoptive transfer of haptenized Cnr2-/- and Cnr1-/-/Cnr2-/- bone marrow (BM) DCs as compared to transfer of WT DCs. In contrast, induction of CHS was not affected in WT recipients after transfer of Cnr1-/- DCs. In vitro stimulated Cnr2-/- DCs showed lower CCR7 and CXCR4 expression when compared to WT cells, while in vitro migration towards the chemokine ligands was not affected by CB2. Upregulation of MHC class II and co-stimulatory molecules was also reduced in Cnr2-/- DCs. This study demonstrates that CB2 modulates the maturation phenotype of DCs but not their chemotactic capacities in vitro. These findings and the fact that CHS responses mediated by Cnr2-/- DCs are reduced suggest that CB2 is a promising target for the treatment of inflammatory skin conditions.


Subject(s)
Dendritic Cells/immunology , Dermatitis, Allergic Contact/immunology , Receptor, Cannabinoid, CB2/metabolism , Animals , Cell Differentiation , Cells, Cultured , Chemotaxis , Dendritic Cells/cytology , Dermatitis, Allergic Contact/genetics , Dinitrofluorobenzene/toxicity , Histocompatibility Antigens/genetics , Histocompatibility Antigens/metabolism , Mice , Mice, Inbred C57BL , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/genetics , Receptors, CCR4/genetics , Receptors, CCR4/metabolism , Receptors, CCR7/genetics , Receptors, CCR7/metabolism
5.
Biochim Biophys Acta Mol Cell Res ; 1865(9): 1277-1292, 2018 09.
Article in English | MEDLINE | ID: mdl-29932987

ABSTRACT

BACKGROUND: The Ras-homologous GTPase Rac1 plays a key role in the regulation of gene expression, cytoskeleton-associated processes and cell death as well as carcinogenesis and inflammation. Here, we investigated the impact of Rac1 signaling on liver-mediated immune homeostasis. METHODS: We employed a constitutive Alb-Cre-driven rac1 knock-out and a poly I:C-inducible Mx1-Cre-based knock-out model and analyzed cytokine expression profiles in liver and other organs under basal situation and following LPS-induced endotoxemia by flow cytometry, qRT-PCR and immunocytochemistry. RESULTS: Constitutive Alb-Cre-driven rac1 knockout in hepatocytes altered the basal distribution and activation of immune cells in the liver and likewise in kidney and lung. Early systemic alterations in cytokine serum levels following LPS treatment remained unaffected by Rac1. Furthermore, lack of Rac1 in hepatocytes of untreated animals shifted the liver to a chronic inflammatory state, as depicted by an enhanced mRNA expression of marker genes related to activated macrophages. Upon acute LPS-induced endotoxemia, increased IL-10 mRNA expression in the liver of Alb-Cre Rac1-deficient mice provided an anti-inflammatory response. Employing a poly I:C-inducible Mx1-Cre-based rac1 knock-out, which allows a more widespread rac1 deletion in both hepatocytes and non-hepatocytes, we observed substantial differences regarding both basal and LPS-stimulated cytokine expression profiles as compared to the Alb-Cre system. CONCLUSIONS: Rac1-dependent mechanisms in hepatocytes and non-hepatocytes contribute to the maintenance of liver immune homeostasis under basal situation and following LPS-induced endotoxemia. Disturbed Rac1-regulated hepatocyte functions may promote liver damage under pathophysiological situation involving inflammatory stress.


Subject(s)
Endotoxemia/enzymology , Interleukin-10/genetics , Lipopolysaccharides/adverse effects , Liver/immunology , Neuropeptides/genetics , Neuropeptides/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , Animals , Disease Models, Animal , Endotoxemia/chemically induced , Endotoxemia/genetics , Endotoxemia/immunology , Gene Expression Regulation , Gene Knockout Techniques , Immunity , Kidney/immunology , Liver/enzymology , Lung/immunology , Macrophages/metabolism , Mice , Signal Transduction
6.
J Virol ; 92(3)2018 02 01.
Article in English | MEDLINE | ID: mdl-29142134

ABSTRACT

Innate immune activation is essential to mount an effective antiviral response and to prime adaptive immunity. Although a crucial role of CD169+ cells during vesicular stomatitis virus (VSV) infections is increasingly recognized, factors regulating CD169+ cells during viral infections remain unclear. Here, we show that tumor necrosis factor is produced by CD11b+ Ly6C+ Ly6G+ cells following infection with VSV. The absence of TNF or TNF receptor 1 (TNFR1) resulted in reduced numbers of CD169+ cells and in reduced type I interferon (IFN-I) production during VSV infection, with a severe disease outcome. Specifically, TNF triggered RelA translocation into the nuclei of CD169+ cells; this translocation was inhibited when the paracaspase MALT-1 was absent. Consequently, MALT1 deficiency resulted in reduced VSV replication, defective innate immune activation, and development of severe disease. These findings indicate that TNF mediates the maintenance of CD169+ cells and innate and adaptive immune activation during VSV infection.IMPORTANCE Over the last decade, strategically placed CD169+ metallophilic macrophages in the marginal zone of the murine spleen and lymph nodes (LN) have been shown to play a very important role in host defense against viral pathogens. CD169+ macrophages have been shown to activate innate and adaptive immunity via "enforced virus replication," a controlled amplification of virus particles. However, the factors regulating the CD169+ macrophages remain to be studied. In this paper, we show that after vesicular stomatitis virus infection, phagocytes produce tumor necrosis factor (TNF), which signals via TNFR1, and promote enforced virus replication in CD169+ macrophages. Consequently, lack of TNF or TNFR1 resulted in defective immune activation and VSV clearance.


Subject(s)
Interferon Type I/immunology , Macrophages/immunology , Tumor Necrosis Factor-alpha/immunology , Vesicular Stomatitis/immunology , Adaptive Immunity , Animals , Immunity, Innate , Macrophages/virology , Mice , Mice, Inbred C57BL , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Receptors, Tumor Necrosis Factor, Type I/immunology , Sialic Acid Binding Ig-like Lectin 1 , Transcription Factor RelA/metabolism , Vesiculovirus/physiology , Virus Replication
7.
Bioorg Med Chem ; 27(23): 115145, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31648874

ABSTRACT

The therapy of cancer continues to be a challenge aggravated by the evolution of resistance against current medications. As an alternative for the traditional tripartite treatment options of surgery, radiation and chemotherapy, immunotherapy is gaining increasing attention due to the opportunity of more targeted approaches. Promising targets are antigen-presenting cells which drive innate and adaptive immune responses. The discovery and emergence of new drugs and lead structures can be inspired by natural products which comprise many highly bioactive molecules. The development of new drugs based on natural products is hampered by the current lack of guidelines for screening these structures for immune activating compounds. In this work, we describe a phenotypic preclinical screening pipeline for first-line identification of promising natural products using the mouse as a model system. Favorable compounds are defined to be non-toxic to immune target cells, to show direct anti-tumor effects and to be immunostimulatory at the same time. The presented screening pipeline constitutes a useful tool and aims to integrate immune activation in experimental approaches early on in drug discovery. It supports the selection of natural products for later chemical optimization, direct application in in vivo mouse models and clinical trials and promotes the emergence of new innovative drugs for cancer treatment.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antineoplastic Agents/pharmacology , Biological Products/pharmacology , Drug Discovery/methods , Drug Screening Assays, Antitumor/methods , Neoplasms/drug therapy , Adjuvants, Immunologic/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Biological Products/therapeutic use , Cells, Cultured , Disease Models, Animal , Humans , Immunotherapy , Mice , Neoplasms/therapy
8.
Int J Mol Sci ; 20(18)2019 Sep 07.
Article in English | MEDLINE | ID: mdl-31500303

ABSTRACT

Viral infections are associated with increased incidence of severe sepsis. Particularly during the early stages, type I interferons (IFNs) are known mediators of detrimental effects. However, the functional role of early interferon ß (IFNß) and its cellular source during sepsis in the context of preexisting viral infections has not been defined. Using the colon ascendens stent peritonitis (CASP) model, we demonstrate that IFNß-/- and type I IFN receptor (IFNAR1)-/- mice were less susceptible to sepsis after pre-stimulation with the viral mimetic poly(I:C). Wild type (WT) mice treated with poly(I:C) exhibited altered expression patterns of TNF and IL-12p40 during CASP which were dependent on IFNß or IFNAR1, suggesting a mechanism for the increased sepsis susceptibility of WT mice. Using a double cytokine reporter mouse model, we present novel data on the simultaneous expression of IFNß and IL-12p40 on a single cell level during polymicrobial sepsis in vivo. Conventional dendritic cells (cDCs) were identified as primary source of IFNß and the protective cytokine IL-12p40 after CASP surgery irrespective of poly(I:C) pre-stimulation. These data demonstrated that if polymicrobial sepsis is preceded by a viral infection, IFNß and IL-12p40 are expressed by polyfunctional cDCs suggesting that these cells can play both detrimental and beneficial roles during sepsis development.


Subject(s)
Coinfection/immunology , Dendritic Cells/immunology , Interferon-beta/genetics , Poly I-C/administration & dosage , Receptor, Interferon alpha-beta/genetics , Sepsis/immunology , Animals , Coinfection/blood , Coinfection/virology , Disease Models, Animal , Female , Gene Knockout Techniques , Interferon-beta/metabolism , Interleukin-12 Subunit p40/metabolism , Mice , Mice, Inbred C57BL , Poly I-C/immunology , Receptor, Interferon alpha-beta/metabolism , Sepsis/virology , Signal Transduction
9.
Int J Mol Sci ; 20(1)2019 Jan 07.
Article in English | MEDLINE | ID: mdl-30621022

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) leading to demyelination and axonal damage. It often affects young adults and can lead to neurological disability. Interferon ß (IFNß) preparations represent widely used treatment regimens for patients with relapsing-remitting MS (RRMS) with therapeutic efficacy in reducing disease progression and frequency of acute exacerbations. In mice, IFNß therapy has been shown to ameliorate experimental autoimmune encephalomyelitis (EAE), an animal model of MS while genetic deletion of IFNß or its receptor augments clinical severity of disease. However, the complex mechanism of action of IFNß in CNS autoimmunity has not been fully elucidated. Here, we review our current understanding of the origin, phenotype, and function of microglia and CNS immigrating macrophages in the pathogenesis of MS and EAE. In addition, we highlight the emerging roles of microglia as IFNß-producing cells and vice versa the impact of IFNß on microglia in CNS autoimmunity. We finally discuss recent progress in unraveling the underlying molecular mechanisms of IFNß-mediated effects in EAE.


Subject(s)
Autoimmunity , Central Nervous System/immunology , Interferon-beta/metabolism , Microglia/metabolism , Neuroprotective Agents/metabolism , Animals , Disease Models, Animal , Humans
10.
Int J Mol Sci ; 20(14)2019 Jul 17.
Article in English | MEDLINE | ID: mdl-31319604

ABSTRACT

Altered adaptive immunity involving T lymphocytes has been found in depressed patients and in stress-induced depression-like behavior in animal models. Peripheral T cells play important roles in homeostasis and function of the central nervous system and thus modulate behavior. However, the T cell phenotype and function associated with susceptibility and resilience to depression remain largely unknown. Here, we characterized splenic T cells in susceptible and resilient mice after 10 days of social defeat stress (SDS). We found equally decreased T cell frequencies and comparably altered expression levels of genes associated with T helper (Th) cell function in resilient and susceptible mice. Interleukin (IL)-17 producing CD4+ and CD8+ T cell numbers in the spleen were significantly increased in susceptible mice. These animals further exhibited significantly reduced numbers of regulatory T cells (Treg) and decreased gene expression levels of TGF-ß. Mice with enhanced Th17 differentiation induced by conditional deletion of PPARγ in CD4+ cells (CD4-PPARγKO), an inhibitor of Th17 development, were equally susceptible to SDS when compared to CD4-PPARγWT controls. These data indicate that enhanced Th17 differentiation alone does not alter stress vulnerability. Thus, SDS promotes Th17 cell and suppresses Treg cell differentiation predominantly in susceptible mice with yet unknown effects in immune responses after stress exposure.


Subject(s)
Cell Differentiation/immunology , Stress, Psychological/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Disease Susceptibility , Male , Mice , Stress, Psychological/pathology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/pathology
11.
BMC Genomics ; 19(1): 194, 2018 Mar 12.
Article in English | MEDLINE | ID: mdl-29703139

ABSTRACT

BACKGROUND: The murine discs large homolog 2 (DLG2; post synaptic density 93 (PSD-93); Chapsyn-110) is a member of the membrane-associated guanylate kinase (MAGUK) protein family involved in receptor assembly and associated with signaling enzymes on cell membranes. In neurons, DLG2 protein isoforms derived from alternatively spliced transcripts have been described to bind to NMDA (N-methyl-aspartate) receptors and K channels and to mediate clustering of these channels in the postsynaptic membrane. In myeloid cells of the immune system, such as dendritic cells (DCs), a lack of data exists on the expression or function of DLG2. In cDNA microarray transcriptome analyses, we found Dlg2 highly expressed in a subpopulation of plasmacytoid DCs (pDCs) stimulated to produce type I interferons (IFNs) such as IFNß. RESULTS: Using RACE- and RT-PCR as well as immunoprecipitation followed by Western blotting we characterised the differential expression of the Dlg2 splice variants in IFNß-producing pDCs. Besides Dlg2É£ this cell population expressed a novel short Dlg2η transcript we termed Dlg2η3. Our expression data were integrated into information from genome databases to obtain a novel and comprehensive overview of the mouse Dlg2 gene architecture. To elucidate the intracellular localisation pattern of protein isoforms, ectopical expression analysis of fluorescently tagged DLG2 splice variants was performed. Here we found an enrichment of the larger isoform DLG2α1 at the plasma membrane while the newly identified shorter (DLG2η) isoform as well as DLG2É£ were equally distributed throughout the cytoplasm. Additionally, DLG2η was also found in the nucleus. Analysis of Dlg2-knockout mice previously generated by deleting exon 9 surprisingly revealed that the protein for the novel DLG2η isoform was still expressed in the brain and in bone marrow-derived pDCs from mice carrying the homozygous deletion (Dlg2 ΔE9/ΔE9 ). CONCLUSION: We describe a novel splice variant of the mouse Dlg2 gene termed Dlg2η and define the differential expression pattern of DLG2 isoforms in IFNß-producing pDCs. The presence of DLG2η protein in the CNS of Dlg2 ΔE9/ΔE9 mice might influence the phenotype of these mice and has to be taken into account in the interpretation of results regarding the functional role of DLG2 in neuronal postsynaptic membranes.


Subject(s)
Alternative Splicing , Dendritic Cells/metabolism , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Interferon-beta/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Animals , Bone Marrow/metabolism , Brain/metabolism , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Dendritic Cells/cytology , Gene Expression Profiling , Gene Knockout Techniques , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , Oligonucleotide Array Sequence Analysis , Protein Isoforms/metabolism , Up-Regulation
12.
J Immunol ; 196(11): 4447-51, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27183572

ABSTRACT

Type I IFNs are critical in initiating protective antiviral immune responses, and plasmacytoid dendritic cells (pDCs) represent a major source of these cytokines. We show that only few pDCs are capable of producing IFN-ß after virus infection or CpG stimulation. Using IFNß/YFP reporter mice, we identify these IFN-ß-producing cells in the spleen as a CCR9(+)CD9(-) pDC subset that is localized exclusively within the T/B cell zones. IFN-ß-producing pDCs exhibit a distinct transcriptome profile, with higher expression of genes encoding cytokines and chemokines, facilitating T cell recruitment and activation. These distinctive characteristics of IFN-ß-producing pDCs are independent of the type I IFNR-mediated feedback loop. Furthermore, IFN-ß-producing pDCs exhibit enhanced CCR7-dependent migratory properties in vitro. Additionally, they effectively recruit T cells in vivo in a peritoneal inflammation model. We define "professional type I IFN-producing cells" as a distinct subset of pDCs specialized in coordinating cellular immune responses.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Interferon-beta/genetics , Spleen/cytology , Spleen/immunology , Transcriptome , Animals , Interferon-beta/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/immunology , Signal Transduction
13.
J Immunol ; 196(6): 2439-43, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26819202

ABSTRACT

Protective immunity against intracellular pathogens involves the induction of robust CTL responses. Vaccination with protein Ags establishes such responses only when combined with immune-stimulatory adjuvants. In this study, we compared different adjuvants and identified triphosphate RNA (3pRNA) as especially effective at inducing CTL responses. 3pRNA sensing required IPS-1/MAVS signaling and induced type I IFN in plasmacytoid dendritic cells and macrophages, with the latter being more important for the adjuvant effect. Type I IFN acted on CD11c(+) cells, especially on CD8α(+) Batf3-dependent dendritic cells. Vaccination with OVA in combination with 3pRNA protected mice from a subsequent OVA-encoding adenovirus infection in a CD8(+) cell-dependent manner and more efficiently than other adjuvants. In summary, 3pRNA is a superior adjuvant for CTL activation and might be useful to facilitate antiviral immunization strategies.


Subject(s)
Cross-Priming/immunology , DEAD-box RNA Helicases/immunology , RNA/immunology , T-Lymphocytes, Cytotoxic/immunology , Viral Vaccines/immunology , Adaptor Proteins, Signal Transducing/immunology , Adjuvants, Immunologic/pharmacology , Animals , DEAD Box Protein 58 , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Ligands , Mice , RNA/pharmacology , Signal Transduction/immunology
14.
J Biol Chem ; 291(37): 19517-31, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27474745

ABSTRACT

Cerebral malaria is a severe and often fatal complication of Plasmodium falciparum infection. It is characterized by parasite sequestration, a breakdown of the blood-brain barrier, and a strong inflammation in the brain. We investigated the role of the cannabinoid receptor 2 (CB2), an important modulator of neuroinflammatory responses, in experimental cerebral malaria (ECM). Strikingly, mice with a deletion of the CB2-encoding gene (Cnr2(-/-)) inoculated with Plasmodium berghei ANKA erythrocytes exhibited enhanced survival and a diminished blood-brain barrier disruption. Therapeutic application of a specific CB2 antagonist also conferred increased ECM resistance in wild type mice. Hematopoietic derived immune cells were responsible for the enhanced protection in bone marrow (BM) chimeric Cnr2(-/-) mice. Mixed BM chimeras further revealed that CB2-expressing cells contributed to ECM development. A heterogeneous CD11b(+) cell population, containing macrophages and neutrophils, expanded in the Cnr2(-/-) spleen after infection and expressed macrophage mannose receptors, arginase-1 activity, and IL-10. Also in the Cnr2(-/-) brain, CD11b(+) cells that expressed selected anti-inflammatory markers accumulated, and expression of inflammatory mediators IFN-γ and TNF-α was reduced. Finally, the M2 macrophage chemokine CCL17 was identified as an essential factor for enhanced survival in the absence of CB2, because CCL17 × Cnr2 double-deficient mice were fully susceptible to ECM. Thus, targeting CB2 may be promising for the development of alternative treatment regimes of ECM.


Subject(s)
Blood-Brain Barrier/immunology , Chemokine CCL17/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Receptor, Cannabinoid, CB2/immunology , Animals , Arginase/genetics , Arginase/immunology , Blood-Brain Barrier/parasitology , Blood-Brain Barrier/pathology , Chemokine CCL17/genetics , Disease Models, Animal , Disease Susceptibility , Female , Interleukin-10/genetics , Interleukin-10/immunology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Macrophages/immunology , Macrophages/pathology , Malaria, Cerebral/genetics , Malaria, Cerebral/pathology , Male , Mannose Receptor , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/immunology , Mice , Mice, Knockout , Neutrophils/immunology , Neutrophils/pathology , Receptor, Cannabinoid, CB2/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology
15.
J Virol ; 90(20): 9330-7, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27512061

ABSTRACT

UNLABELLED: Type I interferons (IFNs) crucially contribute to host survival upon viral infections. Robust expression of type I IFNs (IFN-α/ß) and induction of an antiviral state critically depend on amplification of the IFN signal via the type I IFN receptor (IFNAR). A small amount of type I IFN produced early upon virus infection binds the IFNAR and activates a self-enhancing positive feedback loop, resulting in induction of large, protective amounts of IFN-α. Unexpectedly, we found robust, systemic IFN-α expression upon infection of IFNAR knockout mice with the orthomyxovirus Thogoto virus (THOV). The IFNAR-independent IFN-α production required in vivo conditions and was not achieved during in vitro infection. Using replication-incompetent THOV-derived virus-like particles, we demonstrate that IFNAR-independent type I IFN induction depends on viral polymerase activity but is largely independent of viral replication. To discover the cell type responsible for this effect, we used type I IFN reporter mice and identified CD11b(+) F4/80(+) myeloid cells within the peritoneal cavity of infected animals as the main source of IFNAR-independent type I IFN, corresponding to the particular tropism of THOV for this cell type. IMPORTANCE: Type I IFNs are crucial for the survival of a host upon most viral infections, and, moreover, they shape subsequent adaptive immune responses. Production of protective amounts of type I IFN critically depends on the positive feedback amplification via the IFNAR. Unexpectedly, we observed robust IFNAR-independent type I IFN expression upon THOV infection and unraveled molecular mechanisms and determined the tissue and cell type involved. Our data indicate that the host can effectively use alternative pathways to induce type I IFN responses if the classical feedback amplification is not available. Understanding how type I IFN can be produced in large amounts independently of IFNAR-dependent enhancement will identify mechanisms which might contribute to novel therapeutic strategies to fight viral pathogens.


Subject(s)
CD11b Antigen/metabolism , Interferon Type I/metabolism , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Peritoneum/virology , Receptor, Interferon alpha-beta/metabolism , Thogotovirus/metabolism , Animals , Humans , Interferon-alpha/metabolism , Interferon-beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritoneum/metabolism , Signal Transduction/physiology , Virus Replication/physiology
16.
Brain Behav Immun ; 66: 382-393, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28642092

ABSTRACT

The CC chemokine ligand 17 (CCL17) and its cognate CC chemokine receptor 4 (CCR4) are known to control leukocyte migration, maintenance of TH17 cells, and regulatory T cell (Treg) expansion in vivo. In this study we characterized the expression and functional role of CCL17 in the pathogenesis of experimental autoimmune encephalomyelitis (EAE). Using a CCL17/EGFP reporter mouse model, we could show that CCL17 expression in the CNS can be found in a subset of classical dendritic cells (DCs) that immigrate into the CNS during the effector phase of MOG-induced EAE. CCL17 deficient (CCL17-/-) mice exhibited an ameliorated disease course upon MOG-immunization, associated with reduced immigration of IL-17 producing CD4+ T cells and peripheral DCs into the CNS. CCL17-/- DCs further showed equivalent MHC class II and costimulatory molecule expression and an equivalent capacity to secrete IL-23 and induce myelin-reactive TH17 cells when compared to wildtype DCs. In contrast, their transmigration in an in vitro model of the blood-brain barrier was markedly impaired. In addition, peripheral Treg cells were enhanced in CCL17-/- mice at peak of disease pointing towards an immunoregulatory function of CCL17 in EAE. Our study identifies CCL17 as a unique modulator of EAE pathogenesis regulating DC trafficking as well as peripheral Treg cell expansion in EAE. Thus, CCL17 operates at distinct levels and on different cell subsets during immune response in EAE, a property harboring therapeutic potential for the treatment of CNS autoimmunity.


Subject(s)
Chemokine CCL17/metabolism , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Cell Movement , Chemokine CCL17/genetics , Female , Interleukin-23/metabolism , Mice, Inbred C57BL , Mice, Knockout , Spleen/immunology , Spleen/physiopathology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism
17.
Int J Mol Sci ; 18(11)2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29099057

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system (CNS). It affects more than two million people worldwide, mainly young adults, and may lead to progressive neurological disability. Chemokines and their receptors have been shown to play critical roles in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), a murine disease model induced by active immunization with myelin proteins or transfer of encephalitogenic CD4⁺ T cells that recapitulates clinical and neuropathological features of MS. Chemokine ligand-receptor interactions orchestrate leukocyte trafficking and influence multiple pathophysiological cellular processes, including antigen presentation and cytokine production by dendritic cells (DCs). The C-C class chemokines 17 (CCL17) and 22 (CCL22) and their C-C chemokine receptor 4 (CCR4) have been shown to play an important role in homeostasis and inflammatory responses. Here, we provide an overview of the involvement of CCR4 and its ligands in CNS autoimmunity. We review key clinical studies of MS together with experimental studies in animals that have demonstrated functional roles of CCR4, CCL17, and CCL22 in EAE pathogenesis. Finally, we discuss the therapeutic potential of newly developed CCR4 antagonists and a humanized anti-CCR4 antibody for treatment of MS.


Subject(s)
Autoimmune Diseases of the Nervous System/immunology , Autoimmunity , Chemokine CCL17/immunology , Chemokine CCL22/immunology , Receptors, CCR4/immunology , Animals , Autoimmune Diseases of the Nervous System/drug therapy , Autoimmune Diseases of the Nervous System/pathology , Autoimmunity/immunology , Central Nervous System/drug effects , Central Nervous System/immunology , Central Nervous System/pathology , Chemokine CCL17/analysis , Chemokine CCL22/analysis , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Molecular Targeted Therapy/methods , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Receptors, CCR4/analysis
18.
J Virol ; 89(1): 262-74, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25320325

ABSTRACT

UNLABELLED: Skin keratinocytes represent a primary entry site for herpes simplex virus 1 (HSV-1) in vivo. The cellular proteins nectin-1 and herpesvirus entry mediator (HVEM) act as efficient receptors for both serotypes of HSV and are sufficient for disease development mediated by HSV-2 in mice. How HSV-1 enters skin and whether both nectin-1 and HVEM are involved are not known. We addressed the impact of nectin-1 during entry of HSV-1 into murine epidermis and investigated the putative contribution of HVEM. Using ex vivo infection of murine epidermis, we showed that HSV-1 entered the basal keratinocytes of the epidermis very efficiently. In nectin-1-deficient epidermis, entry was strongly reduced. Almost no entry was observed, however, in nectin-1-deficient keratinocytes grown in culture. This observation correlated with the presence of HVEM on the keratinocyte surface in epidermis and with the lack of HVEM expression in nectin-1-deficient primary keratinocytes. Our results suggest that nectin-1 is the primary receptor in epidermis, while HVEM has a more limited role. For primary murine keratinocytes, on which nectin-1 acts as a single receptor, electron microscopy suggested that HSV-1 can enter both by direct fusion with the plasma membrane and via endocytic vesicles. Thus, we concluded that nectin-1 directs internalization into keratinocytes via alternative pathways. In summary, HSV-1 entry into epidermis was shown to strongly depend on the presence of nectin-1, but the restricted presence of HVEM can potentially replace nectin-1 as a receptor, illustrating the flexibility employed by HSV-1 to efficiently invade tissue in vivo. IMPORTANCE: Herpes simplex virus (HSV) can cause a range of diseases in humans, from uncomplicated mucocutaneous lesions to life-threatening infections. The skin is one target tissue of HSV, and the question of how the virus overcomes the protective skin barrier and penetrates into the tissue to reach its receptors is still open. Previous studies analyzing entry into cells grown in vitro revealed nectin-1 and HVEM as HSV receptors. To explore the contributions of nectin-1 and HVEM to entry into a natural target tissue, we established an ex vivo infection model. Using nectin-1- or HVEM-deficient mice, we demonstrated the distinct involvement of nectin-1 and HVEM for HSV-1 entry into epidermis and characterized the internalization pathways. Such advances in understanding the involvement of receptors in tissue are essential preconditions for unraveling HSV invasion of skin, which in turn will allow the development of antiviral reagents.


Subject(s)
Cell Adhesion Molecules/metabolism , Herpesvirus 1, Human/physiology , Host-Pathogen Interactions , Keratinocytes/virology , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Receptors, Virus/metabolism , Virus Internalization , Animals , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Nectins , Skin/virology
19.
J Immunol ; 193(5): 2496-503, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25070849

ABSTRACT

Upon treatment with vesicular stomatitis virus (VSV) particles, plasmacytoid dendritic cells (pDC) are triggered to mount substantial type I IFN responses, whereas myeloid DC (mDC) are only minor producers. Interestingly, bone marrow-derived (BM-)mDC were more vulnerable to infection with enhanced GFP (eGFP)-expressing VSV (VSVeGFP) than BM-pDC. BM-pDC stimulated with wild-type VSV mounted TLR-dependent IFN responses that were independent of RIG-I-like helicase (RLH) signaling. In contrast, in BM-pDC the VSV variant M2 induced particularly high IFN responses triggered in a TLR- and RLH-dependent manner, whereas BM-mDC stimulation was solely RLH-dependent. Importantly, VSVeGFP treatment of BM-pDC derived from IFN-ß yellow fluorescent protein (YFP) reporter mice (messenger of IFN-ß) resulted in YFP(+) and eGFP(+) single-positive cells, whereas among messenger of IFN-ß-BM-mDC most YFP(+) cells were also eGFP(+). This observation indicated that unlike mDC, direct virus infection was not required to trigger IFN responses of pDC. VSV-infected BM-mDC triggered BM-pDC to mount significantly higher IFN responses than free virus particles. Stimulation with infected cells enhanced the percentages of pDC subsets expressing either IFN-ß(+) or IFN-α6(+) plus IFN-ß(+). Irrespective of whether stimulated with free virus or infected cells, IFN induction was dependent on autophagy of pDC, whereas autophagy of the infected mDC was dispensable. Collectively, these results indicated that productive VSV infection was needed to trigger IFN responses of mDC, but not of pDC, and that IFN responses were primarily induced by virus-infected cells that stimulated pDC in a TLR-dependent manner.


Subject(s)
Dendritic Cells/immunology , Interferon-alpha/immunology , Interferon-beta/immunology , Plasma Cells/immunology , Vesicular Stomatitis/immunology , Vesicular stomatitis Indiana virus/immunology , Animals , Dendritic Cells/pathology , Interferon-alpha/genetics , Interferon-beta/genetics , Mice , Mice, Knockout , Plasma Cells/pathology , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Vesicular Stomatitis/genetics , Vesicular Stomatitis/pathology
20.
J Virol ; 88(23): 13638-50, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25231302

ABSTRACT

UNLABELLED: In healthy individuals, the functional immune system effectively confines human cytomegalovirus (CMV) replication, while viral immune evasion and persistence preclude sterile immunity. Mouse CMV (MCMV) is a well-established model to study the delicate CMV-host balance. Effective control of MCMV infection depends on the induction of protective type I interferon (IFN-I) responses. Nevertheless, it is unclear whether in professional antigen-presenting cell subsets MCMV-encoded evasins inhibit the induction of IFN-I responses. Upon MCMV treatment, enhanced expression of MCMV immediate-early and early proteins was detected in bone marrow cultures of macrophages and myeloid dendritic cells compared with plasmacytoid dendritic cell cultures, whereas plasmacytoid dendritic cells mounted more vigorous IFN-I responses. Experiments with Toll-like receptor (TLR)- and/or RIG-I like helicase (RLH)-deficient cell subsets revealed that upon MCMV treatment of myeloid cells, IFN-I responses were triggered independently of TLR and RLH signaling, whereas in plasmacytoid dendritic cells, IFN-I induction was strictly TLR dependent. Macrophages and myeloid dendritic cells treated with either UV-inactivated MCMV or live MCMV that lacked the STAT2 antagonist M27 mounted significantly higher IFN-I responses than cells treated with live wild-type MCMV. In contrast, plasmacytoid dendritic cells responded similarly to UV-inactivated and live MCMV. These experiments illustrated that M27 not only inhibited IFN-I-mediated receptor signaling, but also evaded the induction of IFN responses in myeloid dendritic cells. Furthermore, we found that additional MCMV-encoded evasins were needed to efficiently shut off IFN-I responses of macrophages, but not of myeloid dendritic cells, thus further elucidating the subtle adjustment of the host-pathogen balance. IMPORTANCE: MCMV may induce IFN-I responses in fibroblasts and epithelial cells, as well as in antigen-presenting cell subsets. We focused on the analysis of IFN-I responses of antigen-presenting cell subsets, including plasmacytoid dendritic cells, myeloid dendritic cells, and macrophages, which are all triggered by MCMV to mount IFN-I responses. Interestingly, myeloid dendritic cells and macrophages, but not plasmacytoid dendritic cells, are readily MCMV infected and support viral gene expression. As expected from previous studies, plasmacytoid dendritic cells sense MCMV Toll-like receptor 9 (TLR9) dependently, whereas in myeloid cells, IFN-I induction is entirely TLR and RLH independent. MCMV-encoded M27 does not impair the IFN-I induction of plasmacytoid dendritic cells, while in myeloid dendritic cells, it reduces IFN-I responses. In macrophages, M27 plus other, not yet identified evasins profoundly inhibit the induction of IFN-I responses. Collectively, these results illustrate that MCMV has evolved diverse mechanisms to differentially modulate IFN-I responses in single immune cell subsets.


Subject(s)
Dendritic Cells/immunology , Immune Evasion , Interferon Type I/antagonists & inhibitors , Interferon Type I/immunology , Muromegalovirus/immunology , Myeloid Cells/immunology , Viral Proteins/immunology , Animals , Cells, Cultured , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL