Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 25(3): 448-461, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38351322

ABSTRACT

Conventional dendritic cells (cDCs) include functionally and phenotypically diverse populations, such as cDC1s and cDC2s. The latter population has been variously subdivided into Notch-dependent cDC2s, KLF4-dependent cDC2s, T-bet+ cDC2As and T-bet- cDC2Bs, but it is unclear how all these subtypes are interrelated and to what degree they represent cell states or cell subsets. All cDCs are derived from bone marrow progenitors called pre-cDCs, which circulate through the blood to colonize peripheral tissues. Here, we identified distinct mouse pre-cDC2 subsets biased to give rise to cDC2As or cDC2Bs. We showed that a Siglec-H+ pre-cDC2A population in the bone marrow preferentially gave rise to Siglec-H- CD8α+ pre-cDC2As in tissues, which differentiated into T-bet+ cDC2As. In contrast, a Siglec-H- fraction of pre-cDCs in the bone marrow and periphery mostly generated T-bet- cDC2Bs, a lineage marked by the expression of LysM. Our results showed that cDC2A versus cDC2B fate specification starts in the bone marrow and suggest that cDC2 subsets are ontogenetically determined lineages, rather than cell states imposed by the peripheral tissue environment.


Subject(s)
Dendritic Cells , Sialic Acid Binding Immunoglobulin-like Lectins , Animals , Mice , Cell Differentiation
2.
Nat Immunol ; 24(4): 714-728, 2023 04.
Article in English | MEDLINE | ID: mdl-36928414

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are the main source of type I interferon (IFN-I) during viral infections. Their other functions are debated, due to a lack of tools to identify and target them in vivo without affecting pDC-like cells and transitional DCs (tDCs), which harbor overlapping phenotypes and transcriptomes but a higher efficacy for T cell activation. In the present report, we present a reporter mouse, pDC-Tom, designed through intersectional genetics based on unique Siglech and Pacsin1 coexpression in pDCs. The pDC-Tom mice specifically tagged pDCs and, on breeding with Zbtb46GFP mice, enabled transcriptomic profiling of all splenic DC types, unraveling diverging activation of pDC-like cells versus tDCs during a viral infection. The pDC-Tom mice also revealed initially similar but later divergent microanatomical relocation of splenic IFN+ versus IFN- pDCs during infection. The mouse models and specific gene modules we report here will be useful to delineate the physiological functions of pDCs versus other DC types.


Subject(s)
Dendritic Cells , Interferon Type I , Animals , Mice , Interferon Type I/metabolism , Gene Expression Profiling , Phenotype , Transcriptome
3.
Nat Immunol ; 21(9): 983-997, 2020 09.
Article in English | MEDLINE | ID: mdl-32690951

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are a major source of type I interferon (IFN-I). What other functions pDCs exert in vivo during viral infections is controversial, and more studies are needed to understand their orchestration. In the present study, we characterize in depth and link pDC activation states in animals infected by mouse cytomegalovirus by combining Ifnb1 reporter mice with flow cytometry, single-cell RNA sequencing, confocal microscopy and a cognate CD4 T cell activation assay. We show that IFN-I production and T cell activation were performed by the same pDC, but these occurred sequentially in time and in different micro-anatomical locations. In addition, we show that pDC commitment to IFN-I production was marked early on by their downregulation of leukemia inhibitory factor receptor and was promoted by cell-intrinsic tumor necrosis factor signaling. We propose a new model for how individual pDCs are endowed to exert different functions in vivo during a viral infection, in a manner tightly orchestrated in time and space.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Herpesviridae Infections/immunology , Muromegalovirus/physiology , Animals , Cells, Cultured , Interferon Type I/metabolism , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Sequence Analysis, RNA , Signal Transduction , Single-Cell Analysis , Tumor Necrosis Factor-alpha/metabolism
4.
Immunity ; 55(11): 2103-2117.e10, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36323311

ABSTRACT

The surface of the central nervous system (CNS) is protected by the meninges, which contain a dense network of meningeal macrophages (MMs). Here, we examined the role of tissue-resident MM in viral infection. MHC-II- MM were abundant neonatally, whereas MHC-II+ MM appeared over time. These barrier macrophages differentially responded to in vivo peripheral challenges such as LPS, SARS-CoV-2, and lymphocytic choriomeningitis virus (LCMV). Peripheral LCMV infection, which was asymptomatic, led to a transient infection and activation of the meninges. Mice lacking macrophages but conserving brain microglia, or mice bearing macrophage-specific deletion of Stat1 or Ifnar, exhibited extensive viral spread into the CNS. Transcranial pharmacological depletion strategies targeting MM locally resulted in several areas of the meninges becoming infected and fatal meningitis. Low numbers of MHC-II+ MM, which is seen upon LPS challenge or in neonates, corelated with higher viral load upon infection. Thus, MMs protect against viral infection and may present targets for therapeutic manipulation.


Subject(s)
COVID-19 , Lymphocytic Choriomeningitis , Animals , Mice , Lipopolysaccharides , Mice, Inbred C57BL , SARS-CoV-2 , Lymphocytic choriomeningitis virus/physiology , Macrophages , Meninges
5.
Immunity ; 47(2): 349-362.e5, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28801233

ABSTRACT

In lymph nodes (LNs), dendritic cells (DCs) are thought to dispose of apoptotic cells, a function pertaining to macrophages in other tissues. We found that a population of CX3CR1+ MERTK+ cells located in the T cell zone of LNs, previously identified as DCs, are efferocytic macrophages. Lineage-tracing experiments and shield chimeras indicated that these T zone macrophages (TZM) are long-lived macrophages seeded in utero and slowly replaced by blood monocytes after birth. Imaging the LNs of mice in which TZM and DCs express different fluorescent proteins revealed that TZM-and not DCs-act as the only professional scavengers, clearing apoptotic cells in the LN T cell zone in a CX3CR1-dependent manner. Furthermore, similar to other macrophages, TZM appear inefficient in priming CD4 T cells. Thus, efferocytosis and T cell activation in the LN are uncoupled processes designated to macrophages and DCs, respectively, with implications to the maintenance of immune homeostasis.


Subject(s)
Lymph Nodes/immunology , Macrophages/immunology , Phagocytosis , Animals , Antigen Presentation , Apoptosis , CD4-Positive T-Lymphocytes/immunology , CX3C Chemokine Receptor 1 , Cell Differentiation , Cell Lineage , Cells, Cultured , Dendritic Cells/immunology , Immune Tolerance , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Chemokine/metabolism , c-Mer Tyrosine Kinase
6.
Immunity ; 47(4): 680-696.e8, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29045900

ABSTRACT

The classical model of hematopoiesis established in the mouse postulates that lymphoid cells originate from a founder population of common lymphoid progenitors. Here, using a modeling approach in humanized mice, we showed that human lymphoid development stemmed from distinct populations of CD127- and CD127+ early lymphoid progenitors (ELPs). Combining molecular analyses with in vitro and in vivo functional assays, we demonstrated that CD127- and CD127+ ELPs emerged independently from lympho-mono-dendritic progenitors, responded differently to Notch1 signals, underwent divergent modes of lineage restriction, and displayed both common and specific differentiation potentials. Whereas CD127- ELPs comprised precursors of T cells, marginal zone B cells, and natural killer (NK) and innate lymphoid cells (ILCs), CD127+ ELPs supported production of all NK cell, ILC, and B cell populations but lacked T potential. On the basis of these results, we propose a "two-family" model of human lymphoid development that differs from the prevailing model of hematopoiesis.


Subject(s)
B-Lymphocytes/metabolism , Killer Cells, Natural/metabolism , Lymphoid Progenitor Cells/metabolism , Lymphopoiesis/genetics , T-Lymphocytes/metabolism , Adolescent , Adult , Animals , B-Lymphocytes/cytology , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , Female , Gene Expression Profiling/methods , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Interleukin-7 Receptor alpha Subunit/genetics , Interleukin-7 Receptor alpha Subunit/metabolism , Killer Cells, Natural/cytology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/transplantation , Male , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Middle Aged , Stem Cell Transplantation , T-Lymphocytes/cytology , Transplantation, Heterologous , Young Adult
7.
Immunity ; 45(2): 305-18, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27533013

ABSTRACT

Dendritic cells (DCs) are instrumental in the initiation of T cell responses, but how thymic and peripheral tolerogenic DCs differ globally from Toll-like receptor (TLR)-induced immunogenic DCs remains unclear. Here, we show that thymic XCR1(+) DCs undergo a high rate of maturation, accompanied by profound gene-expression changes that are essential for central tolerance and also happen in germ-free mice. Those changes largely overlap those occurring during tolerogenic and, more unexpectedly, TLR-induced maturation of peripheral XCR1(+) DCs, arguing against the commonly held view that tolerogenic DCs undergo incomplete maturation. Interferon-stimulated gene (ISG) expression was among the few discriminators of immunogenic and tolerogenic XCR1(+) DCs. Tolerogenic XCR1(+) thymic DCs were, however, unique in expressing ISGs known to restrain virus replication. Therefore, a broad functional convergence characterizes tolerogenic and immunogenic XCR1(+) DC maturation in the thymus and periphery, maximizing antigen presentation and signal delivery to developing and to conventional and regulatory mature T cells.


Subject(s)
Central Tolerance , Dendritic Cells/immunology , Peripheral Tolerance , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Animals , Antigen Presentation , Cell Differentiation , Cells, Cultured , Interferon Regulatory Factors/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Chemokine/metabolism , Toll-Like Receptors/immunology , Transcriptome , Virus Replication
8.
Immunity ; 45(3): 626-640, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27637148

ABSTRACT

Interferon regulatory factor-8 (IRF8) has been proposed to be essential for development of monocytes, plasmacytoid dendritic cells (pDCs) and type 1 conventional dendritic cells (cDC1s) and remains highly expressed in differentiated DCs. Transcription factors that are required to maintain the identity of terminally differentiated cells are designated "terminal selectors." Using BM chimeras, conditional Irf8(fl/fl) mice and various promotors to target Cre recombinase to different stages of monocyte and DC development, we have identified IRF8 as a terminal selector of the cDC1 lineage controlling survival. In monocytes, IRF8 was necessary during early but not late development. Complete or late deletion of IRF8 had no effect on pDC development or survival but altered their phenotype and gene-expression profile leading to increased T cell stimulatory function but decreased type 1 interferon production. Thus, IRF8 differentially controls the survival and function of terminally differentiated monocytes, cDC1s, and pDCs.


Subject(s)
Cell Differentiation/physiology , Dendritic Cells/metabolism , Dendritic Cells/physiology , Interferon Regulatory Factors/metabolism , Transcription Factors/metabolism , Animals , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Monocytes/physiology , Promoter Regions, Genetic/physiology , T-Lymphocytes/metabolism , T-Lymphocytes/physiology
9.
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
10.
Immunity ; 42(4): 627-39, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25862089

ABSTRACT

Migratory non-lymphoid tissue dendritic cells (NLT-DCs) transport antigens to lymph nodes (LNs) and are required for protective immune responses in the context of inflammation and to promote tolerance to self-antigens in steady-state. However, the molecular mechanisms that elicit steady-state NLT-DC maturation and migration are unknown. By comparing the transcriptome of NLT-DCs in the skin with their migratory counterparts in draining LNs, we have identified a novel NF-κB-regulated gene network specific to migratory DCs. We show that targeted deletion of IKKß in DCs, a major activator of NF-κB, prevents NLT-DC accumulation in LNs and compromises regulatory T cell conversion in vivo. This was associated with impaired tolerance and autoimmunity. NF-κB is generally considered the prototypical pro-inflammatory transcription factor, but this study describes a role for NF-κB signaling in DCs for immune homeostasis and tolerance that could have implications in autoimmune diseases and immunity.


Subject(s)
Dendritic Cells/immunology , Gene Regulatory Networks/immunology , Homeostasis/immunology , Immune Tolerance , NF-kappa B/immunology , Signal Transduction/immunology , Animals , Autoantigens/genetics , Autoantigens/immunology , Autoimmunity , Cell Movement , Dendritic Cells/cytology , Gene Expression Profiling , Gene Expression Regulation , I-kappa B Kinase/deficiency , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Knockout , Microarray Analysis , NF-kappa B/genetics , Skin/cytology , Skin/immunology , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
11.
J Cell Sci ; 134(18)2021 09 15.
Article in English | MEDLINE | ID: mdl-34494097

ABSTRACT

Lysosomal signaling facilitates the migration of immune cells by releasing Ca2+ to activate the actin-based motor myosin II at the cell rear. However, how the actomyosin cytoskeleton physically associates to lysosomes is unknown. We have previously identified myosin II as a direct interactor of Rab7b, a small GTPase that mediates the transport from late endosomes/lysosomes to the trans-Golgi network (TGN). Here, we show that Rab7b regulates the migration of dendritic cells (DCs) in one- and three-dimensional environments. DCs are immune sentinels that transport antigens from peripheral tissues to lymph nodes to activate T lymphocytes and initiate adaptive immune responses. We found that the lack of Rab7b reduces myosin II light chain phosphorylation and the activation of the transcription factor EB (TFEB), which controls lysosomal signaling and is required for fast DC migration. Furthermore, we demonstrate that Rab7b interacts with the lysosomal Ca2+ channel TRPML1 (also known as MCOLN1), enabling the local activation of myosin II at the cell rear. Taken together, our findings identify Rab7b as the missing physical link between lysosomes and the actomyosin cytoskeleton, allowing control of immune cell migration through lysosomal signaling. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Actomyosin , Lysosomes , Cytoskeleton , Dendritic Cells , Endosomes , Humans
12.
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
13.
EMBO J ; 37(19)2018 10 01.
Article in English | MEDLINE | ID: mdl-30131424

ABSTRACT

Plasmacytoid dendritic cells (pDC) are the major source of type I interferons (IFN-I) during viral infections, in response to triggering of endosomal Toll-like receptors (TLRs) 7 or 9 by viral single-stranded RNA or unmethylated CpG DNA, respectively. Synthetic ligands have been used to disentangle the underlying signaling pathways. The adaptor protein AP3 is necessary to transport molecular complexes of TLRs, synthetic CpG DNA, and MyD88 into endosomal compartments allowing interferon regulatory factor 7 (IRF7) recruitment whose phosphorylation then initiates IFN-I production. High basal expression of IRF7 by pDC and its further enhancement by positive IFN-I feedback signaling appear to be necessary for robust cytokine production. In contrast, we show here that in vivo during mouse cytomegalovirus (MCMV) infection pDC produce high amounts of IFN-I downstream of the TLR9-to-MyD88-to-IRF7 signaling pathway without requiring IFN-I positive feedback, high IRF7 expression, or AP3-driven endosomal routing of TLRs. Hence, the current model of the molecular requirements for professional IFN-I production by pDC, established by using synthetic TLR ligands, does not strictly apply to a physiological viral infection.


Subject(s)
Dendritic Cells/immunology , Herpesviridae Infections/immunology , Interferon Type I/immunology , Muromegalovirus/immunology , Signal Transduction/immunology , Adaptor Protein Complex 3/genetics , Adaptor Protein Complex 3/immunology , Animals , Dendritic Cells/pathology , Endosomes/genetics , Endosomes/immunology , Herpesviridae Infections/genetics , Herpesviridae Infections/pathology , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon Type I/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Signal Transduction/genetics , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology
14.
Immunity ; 38(2): 336-48, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23352235

ABSTRACT

Dendritic cells (DCs) are critical regulators of immune responses. Under noninflammatory conditions, several human DC subsets have been identified. Little is known, however, about the human DC compartment under inflammatory conditions. Here, we characterize a DC population found in human inflammatory fluids that displayed a phenotype distinct from macrophages from the same fluids and from steady-state lymphoid organ and blood DCs. Transcriptome analysis showed that they correspond to a distinct DC subset and share gene signatures with in vitro monocyte-derived DCs. Moreover, human inflammatory DCs, but not inflammatory macrophages, stimulated autologous memory CD4(+) T cells to produce interleukin-17 and induce T helper 17 (Th17) cell differentiation from naive CD4(+) T cells through the selective secretion of Th17 cell-polarizing cytokines. We conclude that inflammatory DCs represent a distinct human DC subset and propose that they are derived from monocytes and are involved in the induction and maintenance of Th17 cell responses.


Subject(s)
Dendritic Cells/pathology , Inflammation/pathology , Interleukin-17/immunology , Macrophages/pathology , Monocytes/pathology , Th17 Cells/pathology , CD4 Antigens/genetics , CD4 Antigens/immunology , Cell Differentiation , Cells, Cultured , Dendritic Cells/immunology , Humans , Immunologic Memory , Inflammation/genetics , Inflammation/immunology , Interleukin-17/biosynthesis , Lymphocyte Activation , Macrophages/immunology , Monocytes/immunology , Organ Specificity , Signal Transduction , Th1-Th2 Balance , Th17 Cells/immunology , Transcriptome/immunology
15.
Immunity ; 39(5): 925-38, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24184057

ABSTRACT

In the skin, the lack of markers permitting the unambiguous identification of macrophages and of conventional and monocyte-derived dendritic cells (DCs) complicates understanding of their contribution to skin integrity and to immune responses. By combining CD64 and CCR2 staining, we successfully identified each of these cell types and studied their origin, transcriptomic signatures, and migratory and T cell stimulatory properties. We also analyzed the impact of microbiota on their development and their contribution to skin inflammation during contact hypersensitivity. Dermal macrophages had a unique scavenging role and were unable to migrate and activate T cells. Conventional dermal DCs excelled both at migrating and activating T cells. In the steady-state dermis, monocyte-derived DCs are continuously generated by extravasated Ly-6C(hi) monocytes. Their T cell stimulatory capacity combined with their poor migratory ability made them particularly suited to activate skin-tropic T cells. Therefore, a high degree of functional specialization occurs among the mononuclear phagocytes of the skin.


Subject(s)
Dendritic Cells/cytology , Macrophages/cytology , Skin/cytology , Animals , Antigens, Differentiation/analysis , CD11b Antigen/analysis , Cell Lineage , Chemotaxis, Leukocyte , Chromatography, Gel , Dendritic Cells/immunology , Dermatitis, Contact/immunology , Dermatitis, Contact/pathology , Dermis/cytology , Gene Expression Regulation, Developmental , Immunophenotyping/methods , Langerhans Cells/cytology , Langerhans Cells/immunology , Lymphocyte Cooperation , Macrophages/physiology , Mice , Microbiota/immunology , Monocytes/cytology , Principal Component Analysis , Radiation Chimera , Receptors, CCR2/analysis , Receptors, IgG/analysis , Skin/immunology , Skin/microbiology , Specific Pathogen-Free Organisms , Staining and Labeling/methods , Transcriptome
16.
EMBO J ; 36(6): 761-782, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28100675

ABSTRACT

In innate immune responses, induction of type-I interferons (IFNs) prevents virus spreading while viral replication is delayed by protein synthesis inhibition. We asked how cells perform these apparently contradictory activities. Using single fibroblast monitoring by flow cytometry and mathematical modeling, we demonstrate that type-I IFN production is linked to cell's ability to enter dsRNA-activated PKR-dependent translational arrest and then overcome this inhibition by decreasing eIF2α phosphorylation through phosphatase 1c cofactor GADD34 (Ppp1r15a) expression. GADD34 expression, shown here to be dependent on the IRF3 transcription factor, is responsible for a biochemical cycle permitting pulse of IFN synthesis to occur in cells undergoing protein synthesis inhibition. Translation arrest is further demonstrated to be key for anti-viral response by acting synergistically with MAVS activation to amplify TBK1 signaling and IFN-ß mRNA transcription, while GADD34-dependent protein synthesis recovery contributes to the heterogeneous expression of IFN observed in dsRNA-activated cells.


Subject(s)
Gene Expression Regulation , Interferon-beta/metabolism , Protein Biosynthesis , Protein Phosphatase 1/metabolism , RNA, Double-Stranded/immunology , RNA, Double-Stranded/metabolism , Animals , Cells, Cultured , Fibroblasts/immunology , Fibroblasts/virology , Flow Cytometry , Gene Expression Profiling , Immunity, Innate , Mice , Models, Theoretical
17.
Nat Immunol ; 10(1): 75-82, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19029904

ABSTRACT

NKp46+CD3- natural killer lymphocytes isolated from blood, lymphoid organs, lung, liver and uterus can produce granule-dependent cytotoxicity and interferon-gamma. Here we identify in dermis, gut lamina propria and cryptopatches distinct populations of NKp46+CD3- cells with a diminished capacity to degranulate and produce interferon-gamma. In the gut, expression of the transcription factor RORgammat, which is involved in the development of lymphoid tissue-inducer cells, defined a previously unknown subset of NKp46+CD3- lymphocytes. Unlike RORgammat- lamina propria and dermis natural killer cells, gut RORgammat+NKp46+ cells produced interleukin 22. Our data show that lymphoid tissue-inducer cells and natural killer cells shared unanticipated similarities and emphasize the heterogeneity of NKp46+CD3- cells in innate immunity, lymphoid organization and local tissue repair.


Subject(s)
Dermis/immunology , Intestinal Mucosa/immunology , Natural Killer T-Cells/immunology , Receptors, Retinoic Acid/physiology , Receptors, Thyroid Hormone/physiology , Transcription Factors/physiology , Animals , CD3 Complex/metabolism , Cell Division , Humans , Interferon-gamma/biosynthesis , Interleukins/biosynthesis , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Natural Cytotoxicity Triggering Receptor 1/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3 , Peyer's Patches/immunology , Receptors, Retinoic Acid/genetics , Receptors, Thyroid Hormone/genetics , Transcription Factors/genetics , Interleukin-22
18.
Immunity ; 37(1): 3-5, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22840836

ABSTRACT

In this issue of Immunity, Haniffa et al. (2012) identify the presence of professional cross-presenting human dendritic cells in the skin, the liver, and the lung and also presented comparative genomics to align human and mouse dendritic cell types across tissues.

19.
Proc Natl Acad Sci U S A ; 114(4): E540-E549, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28069953

ABSTRACT

Immune response against pathogens is a tightly regulated process that must ensure microbial control while preserving integrity of the infected organs. Tuberculosis (TB) is a paramount example of a chronic infection in which antimicrobial immunity is protective in the vast majority of infected individuals but can become detrimental if not finely tuned. Here, we report that C-type lectin dendritic cell (DC) immunoreceptor (DCIR), a key component in DC homeostasis, is required to modulate lung inflammation and bacterial burden in TB. DCIR is abundantly expressed in pulmonary lesions in Mycobacterium tuberculosis-infected nonhuman primates during both latent and active disease. In mice, we found that DCIR deficiency impairs STAT1-mediated type I IFN signaling in DCs, leading to increased production of IL-12 and increased differentiation of T lymphocytes toward Th1 during infection. As a consequence, DCIR-deficient mice control M. tuberculosis better than WT animals but also develop more inflammation characterized by an increased production of TNF and inducible NOS (iNOS) in the lungs. Altogether, our results reveal a pathway by which a C-type lectin modulates the equilibrium between infection-driven inflammation and pathogen's control through sustaining type I IFN signaling in DCs.


Subject(s)
Dendritic Cells/immunology , Interferon Type I/immunology , Lectins, C-Type/immunology , Tuberculosis/immunology , Animals , Female , Lectins, C-Type/genetics , Macaca mulatta , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , STAT1 Transcription Factor/immunology , Signal Transduction
20.
J Immunol ; 198(7): 2785-2795, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28228559

ABSTRACT

Targeting Ags to conventional dendritic cells can enhance Ag-specific immune responses. Although most studies have focused on the induction of T cell responses, the mechanisms by which targeting improves Ab responses are poorly understood. In this study we present data on the use of human XCL1 (hXCL1) and hXCL2 fusion vaccines in a murine model. We show that the human chemokines bound type 1 conventional dendritic cells (cDC1), and that immunization with influenza virus hemagglutinin fused to hXCL1 or hXCL2 induced full protection against influenza challenge. Surprisingly, the hXCL1- and hXCL2-fusion vaccines induced better long-term protection associated with stronger induction of neutralizing Abs, and more Ab-secreting cells in bone marrow. In contrast, murine Xcl1 fusion vaccines induced stronger CD8+ T cell responses compared with hXCL1. Further analysis revealed that although murine Xcl1 fusion vaccines induced chemotaxis and were rapidly endocytosed by cDC1, hXCL1 and hXCL2 fusion vaccines did not induce chemotaxis, were less efficiently endocytosed, and consequently, remained on the surface. This difference may explain the enhanced induction of Abs when targeting Ag to cDC1 using hXCL1 and hXCL2, and suggests that immune responses can be manipulated in directing Abs or T cells based on how efficiently the targeted Ag is endocytosed by the DC.


Subject(s)
Dendritic Cells/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/immunology , Receptors, G-Protein-Coupled/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Endocytosis/immunology , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Flow Cytometry , Humans , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology
SELECTION OF CITATIONS
SEARCH DETAIL