Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Immunity ; 57(5): 1037-1055.e6, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38593796

ABSTRACT

Memory B cells (MBCs) are key providers of long-lived immunity against infectious disease, yet in chronic viral infection, they do not produce effective protection. How chronic viral infection disrupts MBC development and whether such changes are reversible remain unknown. Through single-cell (sc)ATAC-seq and scRNA-seq during acute versus chronic lymphocytic choriomeningitis viral infection, we identified a memory subset enriched for interferon (IFN)-stimulated genes (ISGs) during chronic infection that was distinct from the T-bet+ subset normally associated with chronic infection. Blockade of IFNAR-1 early in infection transformed the chromatin landscape of chronic MBCs, decreasing accessibility at ISG-inducing transcription factor binding motifs and inducing phenotypic changes in the dominating MBC subset, with a decrease in the ISG subset and an increase in CD11c+CD80+ cells. However, timing was critical, with MBCs resistant to intervention at 4 weeks post-infection. Together, our research identifies a key mechanism to instruct MBC identity during viral infection.


Subject(s)
Epigenesis, Genetic , Interferon Type I , Lymphocytic Choriomeningitis , Lymphocytic choriomeningitis virus , Memory B Cells , Animals , Interferon Type I/metabolism , Interferon Type I/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Mice , Lymphocytic choriomeningitis virus/immunology , Memory B Cells/immunology , Mice, Inbred C57BL , Receptor, Interferon alpha-beta/genetics , Immunologic Memory/immunology , Chronic Disease , B-Lymphocyte Subsets/immunology , Single-Cell Analysis
2.
Nat Immunol ; 19(9): 942-953, 2018 09.
Article in English | MEDLINE | ID: mdl-30111894

ABSTRACT

The sensing of microbial genetic material by leukocytes often elicits beneficial pro-inflammatory cytokines, but dysregulated responses can cause severe pathogenesis. Genome-wide association studies have linked the gene encoding phospholipase D3 (PLD3) to Alzheimer's disease and have linked PLD4 to rheumatoid arthritis and systemic sclerosis. PLD3 and PLD4 are endolysosomal proteins whose functions are obscure. Here, PLD4-deficient mice were found to have an inflammatory disease, marked by elevated levels of interferon-γ (IFN-γ) and splenomegaly. These phenotypes were traced to altered responsiveness of PLD4-deficient dendritic cells to ligands of the single-stranded DNA sensor TLR9. Macrophages from PLD3-deficient mice also had exaggerated TLR9 responses. Although PLD4 and PLD3 were presumed to be phospholipases, we found that they are 5' exonucleases, probably identical to spleen phosphodiesterase, that break down TLR9 ligands. Mice deficient in both PLD3 and PLD4 developed lethal liver inflammation in early life, which indicates that both enzymes are needed to regulate inflammatory cytokine responses via the degradation of nucleic acids.


Subject(s)
Dendritic Cells/physiology , Endosomes/metabolism , Exonucleases/metabolism , Hepatitis/genetics , Macrophages/physiology , Membrane Glycoproteins/metabolism , Phospholipase D/metabolism , Alzheimer Disease/genetics , Animals , Arthritis, Rheumatoid/genetics , DNA, Single-Stranded/immunology , Exonucleases/genetics , Genome-Wide Association Study , Humans , Interferon-gamma/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phospholipase D/genetics , Scleroderma, Systemic/genetics , Signal Transduction , Toll-Like Receptor 9/metabolism
3.
Immunity ; 54(6): 1338-1351.e9, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33862015

ABSTRACT

Despite advances in single-cell multi-omics, a single stem or progenitor cell can only be tested once. We developed clonal multi-omics, in which daughters of a clone act as surrogates of the founder, thereby allowing multiple independent assays per clone. With SIS-seq, clonal siblings in parallel "sister" assays are examined either for gene expression by RNA sequencing (RNA-seq) or for fate in culture. We identified, and then validated using CRISPR, genes that controlled fate bias for different dendritic cell (DC) subtypes. This included Bcor as a suppressor of plasmacytoid DC (pDC) and conventional DC type 2 (cDC2) numbers during Flt3 ligand-mediated emergency DC development. We then developed SIS-skew to examine development of wild-type and Bcor-deficient siblings of the same clone in parallel. We found Bcor restricted clonal expansion, especially for cDC2s, and suppressed clonal fate potential, especially for pDCs. Therefore, SIS-seq and SIS-skew can reveal the molecular and cellular mechanisms governing clonal fate.


Subject(s)
Dendritic Cells/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Cell Lineage/genetics , Female , Gene Expression/genetics , HEK293 Cells , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Stem Cells/metabolism
4.
EMBO J ; 42(12): e112712, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37139896

ABSTRACT

cGAS-STING signalling is induced by detection of foreign or mislocalised host double-stranded (ds)DNA within the cytosol. STING acts as the major signalling hub, where it controls production of type I interferons and inflammatory cytokines. Basally, STING resides on the ER membrane. Following activation STING traffics to the Golgi to initiate downstream signalling and subsequently to endolysosomal compartments for degradation and termination of signalling. While STING is known to be degraded within lysosomes, the mechanisms controlling its delivery remain poorly defined. Here we utilised a proteomics-based approach to assess phosphorylation changes in primary murine macrophages following STING activation. This identified numerous phosphorylation events in proteins involved in intracellular and vesicular transport. We utilised high-temporal microscopy to track STING vesicular transport in live macrophages. We subsequently identified that the endosomal complexes required for transport (ESCRT) pathway detects ubiquitinated STING on vesicles, which facilitates the degradation of STING in murine macrophages. Disruption of ESCRT functionality greatly enhanced STING signalling and cytokine production, thus characterising a mechanism controlling effective termination of STING signalling.


Subject(s)
Immunity, Innate , Membrane Proteins , Mice , Animals , Membrane Proteins/genetics , Membrane Proteins/metabolism , Signal Transduction/physiology , Macrophages/metabolism , Nucleotidyltransferases/metabolism , DNA , Endosomal Sorting Complexes Required for Transport/genetics
5.
Immunity ; 47(3): 498-509.e6, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28916264

ABSTRACT

Double-stranded RNA (dsRNA) is a common by-product of viral infections and acts as a potent trigger of antiviral immunity. In the nematode C. elegans, sid-1 encodes a dsRNA transporter that is highly conserved throughout animal evolution, but the physiological role of SID-1 and its orthologs remains unclear. Here, we show that the mammalian SID-1 ortholog, SIDT2, is required to transport internalized extracellular dsRNA from endocytic compartments into the cytoplasm for immune activation. Sidt2-deficient mice exposed to extracellular dsRNA, encephalomyocarditis virus (EMCV), and herpes simplex virus 1 (HSV-1) show impaired production of antiviral cytokines and-in the case of EMCV and HSV-1-reduced survival. Thus, SIDT2 has retained the dsRNA transport activity of its C. elegans ortholog, and this transport is important for antiviral immunity.


Subject(s)
Immunity, Innate , Membrane Proteins/metabolism , RNA Transport , RNA, Double-Stranded/immunology , RNA, Double-Stranded/metabolism , Animals , Cardiovirus Infections/genetics , Cardiovirus Infections/immunology , Cell Line , Cytoplasm , DEAD Box Protein 58/metabolism , Disease Models, Animal , Encephalomyocarditis virus/genetics , Encephalomyocarditis virus/immunology , Endosomes/metabolism , Female , Gene Expression , Gene Knockout Techniques , Herpes Simplex/genetics , Herpes Simplex/immunology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Lysosomes/metabolism , Membrane Proteins/genetics , Mice , Mice, Knockout , Nucleotide Transport Proteins , Protein Binding , Protein Transport , RNA, Viral/genetics , RNA, Viral/metabolism , Signal Transduction , Toll-Like Receptor 3/metabolism
6.
J Immunol ; 210(3): 297-309, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36524995

ABSTRACT

CD8 virtual memory T (TVM) cells are Ag-naive CD8 T cells that have undergone partial differentiation in response to common γ-chain cytokines, particularly IL-15 and IL-4. TVM cells from young individuals are highly proliferative in response to TCR and cytokine stimulation but, with age, they lose TCR-mediated proliferative capacity and exhibit hallmarks of senescence. Helminth infection can drive an increase in TVM cells, which is associated with improved pathogen clearance during subsequent infectious challenge in young mice. Given the cytokine-dependent profile of TVM cells and their age-associated dysfunction, we traced proliferative and functional changes in TVM cells, compared with true naive CD8 T cells, after helminth infection of young and aged C57BL/6 mice. We show that IL-15 is essential for the helminth-induced increase in TVM cells, which is driven only by proliferation of existing TVM cells, with negligible contribution from true naive cell differentiation. Additionally, TVM cells showed the greatest proliferation in response to helminth infection and IL-15 compared with other CD8 T cells. Furthermore, TVM cells from aged mice did not undergo expansion after helminth infection due to both TVM cell-intrinsic and -extrinsic changes associated with aging.


Subject(s)
Helminthiasis , Interleukin-15 , Animals , Mice , Aging/immunology , CD8-Positive T-Lymphocytes/parasitology , Cytokines , Helminthiasis/immunology , Helminthiasis/metabolism , Helminths/pathogenicity , Immunologic Memory , Interleukin-15/metabolism , Mice, Inbred C57BL , Receptors, Antigen, T-Cell
7.
J Immunol ; 207(7): 1836-1847, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34479944

ABSTRACT

DEC-205 is a cell-surface receptor that transports bound ligands into the endocytic pathway for degradation or release within lysosomal endosomes. This receptor has been reported to bind a number of ligands, including keratin, and some classes of CpG oligodeoxynucleotides (ODN). In this study, we explore in detail the requirements for binding ODNs, revealing that DEC-205 efficiently binds single-stranded, phosphorothioated ODN of ≥14 bases, with preference for the DNA base thymidine, but with no requirement for a CpG motif. DEC-205 fails to bind double-stranded phosphodiester ODN, and thus does not bind the natural type of DNA found in mammals. The ODN binding preferences of DEC-205 result in strong binding of B class ODN, moderate binding to C class ODN, minimal binding to P class ODN, and no binding to A class ODN. Consistent with DEC-205 binding capacity, induction of serum IL-12p70 or activation of B cells by each class of ODN correlated with DEC-205 dependence in mice. Thus, the greater the DEC-205 binding capacity, the greater the dependence on DEC-205 for optimal responses. Finally, by covalently linking a B class ODN that efficiently binds DEC-205, to a P class ODN that shows poor binding, we improved DEC-205 binding and increased adjuvancy of the hybrid ODN. The hybrid ODN efficiently enhanced induction of effector CD8 T cells in a DEC-205-dependent manner. Furthermore, the hybrid ODN induced robust memory responses, and was particularly effective at promoting the development of liver tissue-resident memory T cells.


Subject(s)
Adjuvants, Immunologic , Oligodeoxyribonucleotides , Animals , Dendritic Cells , Interleukin-12 , Liver , Mice
8.
Int J Eat Disord ; 55(5): 664-677, 2022 05.
Article in English | MEDLINE | ID: mdl-35302253

ABSTRACT

OBJECTIVE: Identifying risk factors that contribute to the development of anorexia nervosa (AN) is critical for the implementation of early intervention strategies. Anxiety, obsessive-compulsive behavior, and immune dysfunction may be involved in the development of AN; however, their direct influence on susceptibility to the condition remains unclear. Here, we used the activity-based anorexia (ABA) model to examine whether activity, anxiety-like behavior, compulsive behavior, and circulating immune markers predict the subsequent development of pathological weight loss. METHOD: Female Sprague-Dawley rats (n = 44) underwent behavioral testing before exposure to ABA conditions after which they were separated into susceptible and resistant subpopulations. Blood was sampled before behavioral testing and after recovery from ABA to screen for proinflammatory cytokines. RESULTS: Rats that were vulnerable to pathological weight loss differed significantly from resistant rats on all key ABA parameters. While the primary measures of anxiety-like or compulsive behavior were not shown to predict vulnerability to ABA, increased locomotion and anxiety-like behavior were both associated with the extent of weight loss in susceptible but not resistant animals. Moreover, the change in expression of proinflammatory markers IL-4 and IL-6 evoked by ABA was associated with discrete vulnerability factors. Intriguingly, behavior related to risk assessment was shown to predict vulnerability to ABA. DISCUSSION: We did not find undisputable behavioral or immune predictors of susceptibility to pathological weight loss in the ABA rat model. Future research should examine the role of cognition in the development of ABA, dysfunction of which may represent an endophenotype linking anorectic, anxiety-like and compulsive behavior. PUBLIC SIGNIFICANCE: Anorexia nervosa (AN) has among the highest mortality rates of all psychiatric disorders and treatment options remain limited in their efficacy. Understanding what types of risk factors contribute to the development of AN is essential for implementing early intervention strategies. This study describes how some of the most common psychological features of AN could be used to predict susceptibility to pathological weight loss in a well-established animal model.


Subject(s)
Anorexia Nervosa , Anorexia , Adolescent , Animals , Anorexia/pathology , Anorexia Nervosa/diagnosis , Biomarkers , Disease Models, Animal , Female , Humans , Rats , Rats, Sprague-Dawley , Weight Loss/physiology
9.
Eur J Immunol ; 50(10): 1525-1536, 2020 10.
Article in English | MEDLINE | ID: mdl-32383212

ABSTRACT

Initiation of adaptive immunity to particulate antigens in lymph nodes largely depends on their presentation by migratory dendritic cells (DCs). DC subsets differ in their capacity to induce specific types of immunity, allowing subset-specific DC-targeting to influence vaccination and therapy outcomes. Faithful drug design, however, requires exact understanding of subset-specific versus global activation mechanisms. cDC1, the subset of DCs that excel in supporting immunity toward viruses, intracellular bacteria, and tumors, express uniquely high levels of the pattern recognition receptor TLR3. Using various murine genetic models, we show here that both, the cDC1 and cDC2 subsets of cDCs are activated and migrate equally well in response to TLR3 stimulation in a cell extrinsic and TNF-α dependent manner, but that cDC1 show a unique requirement for type I interferon signaling. Our findings reveal common and differing pathways regulating DC subset migration, offering important insights for the design of DC-based vaccination and therapy approaches.


Subject(s)
Dendritic Cells/immunology , Intestines/immunology , Toll-Like Receptor 3/metabolism , Animals , Cancer Vaccines , Cell Movement , Cells, Cultured , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Toll-Like Receptor 3/immunology
10.
Semin Cell Dev Biol ; 84: 11-21, 2018 12.
Article in English | MEDLINE | ID: mdl-29246859

ABSTRACT

Dendritic cells (DC) are professional antigen presenting cells comprising a variety of subsets, as either resident or migrating cells, in lymphoid and non-lymphoid organs. In the steady state DC continually process and present antigens on MHCI and MHCII, processes that are highly upregulated upon activation. By expressing differential sets of pattern recognition receptors different DC subsets are able to respond to a range of pathogenic and danger stimuli, enabling functional specialisation of the DC. The knowledge of functional specialisation of DC subsets is key to efficient priming of T cells, to the design of effective vaccine adjuvants and to understanding the role of different DC in health and disease. This review outlines mouse and human steady state DC subsets and key attributes that define their distinct functions.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/cytology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Dendritic Cells/immunology , Humans
11.
Immunol Cell Biol ; 98(1): 42-53, 2020 01.
Article in English | MEDLINE | ID: mdl-31559654

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) presents an increasing threat to public health, with antimicrobial resistance on the rise and infections endemic in the hospital setting. Despite a global research effort to understand and combat antimicrobial resistance, less work has focused on understanding the nuances in the immunopathogenesis of clinical strains. In particular, there is a surprising gap of knowledge in the literature pertaining to how clinical strains are recognized by dendritic cells (DCs). Here, we show that the activation of DCs is compromised in response to MRSA strains resistant to the last-line antibiotic daptomycin. We found a significant reduction in the secretion of proinflammatory cytokines including tumor necrosis factor-α, interleukin-6, regulated upon activation, normal T cell expressed, and secreted and macrophage inflammatory protein-1ß, as well as decreased expression of CD80 by DCs responding to daptomycin-resistant MRSA. We further demonstrate that this phenotype is coincident with the acquisition of specific point mutations in the cardiolipin synthase gene cls2, and, partly, in the bifunctional lysylphosphatidylglycerol flippase/synthetase gene mprF, which are genes that are often mutated in clinical daptomycin-resistant strains. Therefore, throughout infection and antibiotic therapy, MRSA has the capacity to not only develop further antibiotic resistance, but also develop resistance to immunological recognition by DCs, because of single amino acid point mutations occurring under the selective pressures of both host immunity and antibiotic therapy. Understanding the diversity of clinical MRSA isolates and the nuances in their immune recognition will have important implications for future therapeutics and the treatment of these infections.


Subject(s)
Daptomycin , Dendritic Cells/immunology , Drug Resistance, Bacterial/immunology , Methicillin-Resistant Staphylococcus aureus/immunology , Animals , B7-1 Antigen/immunology , Cytokines/immunology , Gene Expression Regulation , Humans , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Mice
12.
Immunol Cell Biol ; 98(3): 203-214, 2020 03.
Article in English | MEDLINE | ID: mdl-31916630

ABSTRACT

The NZB/W F1 (F1) mice develop severe disease that is similar to human systemic lupus erythematosus. By contrast, each parent strain, NZB or NZW, has limited autoimmunity, suggesting traits of both strains contribute to pathogenesis. Although many of the contributing genes have been identified, the contributing cellular abnormality associated with each parent strain remains unresolved. Given that plasmacytoid dendritic cells (pDCs) are key to the pathogenesis of lupus, we investigated the properties of pDCs from NZB and NZW mice. We found that NZB mouse had higher numbers of pDCs, with much of the increase being contributed by a more abundant CD8+ pDC subset. This was associated with prolonged survival and stronger proliferation of CD4+ T cells. By contrast, NZW pDCs had heightened capacity to produce interferon-α (IFNα) and IFNλ, and promoted stronger B-cell proliferation upon CpG stimulation. Thus, our data reveal the different functional and numerical characteristics of pDCs from NZW and NZB mouse.


Subject(s)
Autoimmunity , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation/immunology , Animals , B-Lymphocytes/immunology , CD8 Antigens/immunology , CD8 Antigens/metabolism , Cell Survival/immunology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Forkhead Transcription Factors/metabolism , Interferon-alpha/metabolism , Interferons/metabolism , Lupus Erythematosus, Systemic/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NZB , Mice, Knockout , Mice, Transgenic , Oligodeoxyribonucleotides/pharmacology
13.
Nat Immunol ; 9(11): 1244-52, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18849989

ABSTRACT

The importance of conventional dendritic cells (cDCs) in the processing and presentation of antigen is well established, but the contribution of plasmacytoid dendritic cells (pDCs) to these processes, and hence to T cell immunity, remains unclear. Here we showed that unlike cDCs, pDCs continued to synthesize major histocompatibility complex (MHC) class II molecules and the MHC class II ubiquitin ligase MARCH1 long after activation. Sustained MHC class II-peptide complex formation, ubiquitination and turnover rendered pDCs inefficient in the presentation of exogenous antigens but enabled pDCs to continuously present endogenous viral antigens in their activated state. As the antigen-presenting abilities of cDCs and pDCs are fundamentally distinct, these two cell types may activate largely nonoverlapping repertoires of CD4(+) T cells.


Subject(s)
Antigen Presentation , Dendritic Cells/immunology , Histocompatibility Antigens Class II/metabolism , Ubiquitination , Animals , Antigens, Viral/immunology , CD11 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/metabolism , Histocompatibility Antigens Class II/biosynthesis , Leukocyte Common Antigens/metabolism , Lymphocyte Activation , Mice , Mice, Inbred Strains , Mice, Knockout , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
14.
Immunity ; 32(2): 279-89, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20171123

ABSTRACT

Dendritic cell (DC) populations consist of multiple subsets that are essential orchestrators of the immune system. Technological limitations have so far prevented systems-wide accurate proteome comparison of rare cell populations in vivo. Here, we used high-resolution mass spectrometry-based proteomics, combined with label-free quantitation algorithms, to determine the proteome of mouse splenic conventional and plasmacytoid DC subsets to a depth of 5,780 and 6,664 proteins, respectively. We found mutually exclusive expression of pattern recognition pathways not previously known to be different among conventional DC subsets. Our experiments assigned key viral recognition functions to be exclusively expressed in CD4(+) and double-negative DCs. The CD8alpha(+) DCs largely lack the receptors required to sense certain viruses in the cytoplasm. By avoiding activation via cytoplasmic receptors, including retinoic acid-inducible gene I, CD8alpha(+) DCs likely gain a window of opportunity to process and present viral antigens before activation-induced shutdown of antigen presentation pathways occurs.


Subject(s)
DEAD-box RNA Helicases/biosynthesis , Dendritic Cells/metabolism , Proteomics/methods , Respirovirus Infections/immunology , Sendai virus/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Cell Separation , Cells, Cultured , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , Flow Cytometry , Host-Pathogen Interactions , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Proteomics/instrumentation , Sendai virus/pathogenicity
15.
J Immunol ; 198(7): 2649-2660, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28202617

ABSTRACT

Nucleotide-binding and oligomerization domain (NOD)-like receptors NOD1 and NOD2 are cytosolic innate immune receptors that recognize microbial peptidoglycans. Although studies have addressed the role of NOD proteins in innate immune responses, little attention has been given to their impact on the developing adaptive immune system. We have assessed the roles of NOD1 and NOD2 deficiency on T cell development in mice. Our results demonstrate that NOD1 and NOD2 promote the positive selection/maturation of CD8 single-positive thymocytes in a thymocyte-intrinsic manner. TCR-mediated ERK phosphorylation is significantly reduced in the absence of NOD proteins, but receptor-interacting protein 2 is not involved in CD8 single-positive thymocyte selection or ERK signaling. Commensal bacteria-free animals have thymocyte maturation defects, and exogenous NOD ligands can enhance thymocyte maturation in culture. These results raise the intriguing possibility that abnormal lymphocyte responses observed in NOD-dependent inflammatory diseases are not driven solely by microbial signals in the gut, but may also involve intrinsic lymphocyte defects resulting from impaired CD8 T cell thymic development.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Thymocytes/cytology , Animals , CD8-Positive T-Lymphocytes/immunology , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction , Thymocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology
16.
EMBO J ; 31(3): 692-706, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22124325

ABSTRACT

The role of specific members of the NF-κB family of transcription factors in CD8 T-cell selection and development is largely unknown. Here, we show that mice lacking NF-κB1 develop a unique population of conventional CD8 single-positive (SP) thymocytes with memory T cell-like properties that populate peripheral immune organs. Development of this memory-like population is not due to PLZF(+) thymocytes and instead coincides with changes in CD8 T-cell selection. These include a reduction in the efficiency of negative selection and a dependence on MHC class Ia or Ib expressed by haematopoietic cells. These findings indicate that NF-κB1 regulates multiple events in the thymus that collectively inhibit the excess development of CD8(+) thymocytes with memory cell characteristics.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/physiology , NF-kappa B/physiology , Thymus Gland/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Immunophenotyping , Interleukin-4/biosynthesis , NF-kappa B/genetics , Signal Transduction
17.
Immunol Cell Biol ; 94(4): 400-10, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26657145

ABSTRACT

Dendritic cells (DCs) are the key initiators of T-helper (Th) 2 immune responses against the parasitic helminth Schistosoma mansoni. Although the liver is one of the main sites of antigen deposition during infection with this parasite, it is not yet clear how distinct DC subtypes in this tissue respond to S. mansoni antigens in vivo, or how the liver microenvironment might influence DC function during establishment of the Th2 response. In this study, we show that hepatic DC subsets undergo distinct activation processes in vivo following murine infection with S. mansoni. Conventional DCs (cDCs) from schistosome-infected mice upregulated expression of the costimulatory molecule CD40 and were capable of priming naive CD4(+) T cells, whereas plasmacytoid DCs (pDCs) upregulated expression of MHC class II, CD86 and CD40 but were unable to support the expansion of either naive or effector/memory CD4(+) T cells. Importantly, in vivo depletion of pDCs revealed that this subset was dispensable for either maintenance or regulation of the hepatic Th2 effector response during acute S. mansoni infection. Our data provides strong evidence that S. mansoni infection favors the establishment of an immunogenic, rather than tolerogenic, liver microenvironment that conditions cDCs to initiate and maintain Th2 immunity in the context of ongoing antigen exposure.


Subject(s)
Dendritic Cells/immunology , Liver/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Th2 Cells/immunology , Animals , Antigens, Helminth/immunology , Cell Differentiation , Cells, Cultured , Dendritic Cells/parasitology , Liver/parasitology , Lymphocyte Activation , Mice , Mice, Inbred C57BL
18.
J Immunol ; 192(4): 1982-9, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24453245

ABSTRACT

We established a humanized mouse model incorporating FLT3-ligand (FLT3-L) administration after hematopoietic cell reconstitution to investigate expansion, phenotype, and function of human dendritic cells (DC). FLT3-L increased numbers of human CD141(+) DC, CD1c(+) DC, and, to a lesser extent, plasmacytoid DC (pDC) in the blood, spleen, and bone marrow of humanized mice. CD1c(+) DC and CD141(+) DC subsets were expanded to a similar degree in blood and spleen, with a bias toward expansion of the CD1c(+) DC subset in the bone marrow. Importantly, the human DC subsets generated after FLT3-L treatment of humanized mice are phenotypically and functionally similar to their human blood counterparts. CD141(+) DC in humanized mice express C-type lectin-like receptor 9A, XCR1, CADM1, and TLR3 but lack TLR4 and TLR9. They are major producers of IFN-λ in response to polyinosinic-polycytidylic acid but are similar to CD1c(+) DC in their capacity to produce IL-12p70. Although all DC subsets in humanized mice are efficient at presenting peptide to CD8(+) T cells, CD141(+) DC are superior in their capacity to cross-present protein Ag to CD8(+) T cells following activation with polyinosinic-polycytidylic acid. CD141(+) DC can be targeted in vivo following injection of Abs against human DEC-205 or C-type lectin-like receptor 9A. This model provides a feasible and practical approach to dissect the function of human CD141(+) and CD1c(+) DC and evaluate adjuvants and DC-targeting strategies in vivo.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antigens, CD1/metabolism , Antigens, Surface/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Glycoproteins/metabolism , Membrane Proteins/pharmacology , Adjuvants, Immunologic/administration & dosage , Animals , Antigen Presentation/immunology , Antigens, CD/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion Molecule-1 , Cell Adhesion Molecules/metabolism , Female , Humans , Immunoglobulins/metabolism , Interferon-gamma/metabolism , Interleukin-12/metabolism , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Lymphocyte Activation/immunology , Membrane Proteins/administration & dosage , Mice , Mice, Inbred NOD , Mice, SCID , Minor Histocompatibility Antigens , Poly I-C/immunology , Receptors, Cell Surface/immunology , Receptors, Chemokine/metabolism , Thrombomodulin , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/metabolism
19.
Cell Mol Life Sci ; 72(22): 4309-25, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26243730

ABSTRACT

The method of choice for the development of new vaccines is to target distinct dendritic cell subsets with antigen in vivo and to harness their function in situ to enhance cell-mediated immunity or induce tolerance to specific antigens. The innate functions of dendritic cells themselves may also be targeted by inhibitors or activators that would target a specific function such as interferon production, potentially important in autoimmune disease and chronic viral infections. Importantly targeting dendritic cells requires detailed knowledge of both the surface phenotype and function of each dendritic cell subset, including how they may respond to different types of vaccine adjuvants, their ability to produce soluble mediators and to process and present antigens and induce priming of naïve T cells. This review summarizes our knowledge of the functional attributes of the human dendritic cell subsets in the steady state and upon activation and their roles in human disease.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Immune System Diseases/immunology , Immunity, Cellular/immunology , T-Lymphocytes/immunology , Antigens, CD1/immunology , Antigens, CD1/metabolism , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/metabolism , Glycoproteins/immunology , Glycoproteins/metabolism , Humans , Infections/immunology , Lymphocyte Activation/immunology
20.
J Virol ; 88(2): 799-810, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24227841

ABSTRACT

RNA-specific adenosine deaminase (ADAR)-mediated adenosine-to-inosine (A-to-I) editing is a critical arm of the antiviral response. However, mechanistic insights into how A-to-I RNA editing affects viral infection are lacking. We posited that inosine incorporation into RNA facilitates sensing of nonself RNA by innate immune sensors and accordingly investigated the impact of inosine-modified RNA on Toll-like receptor 7 and 8 (TLR7/8) sensing. Inosine incorporation into synthetic single-stranded RNA (ssRNA) potentiated tumor necrosis factor alpha (TNF-α) or alpha interferon (IFN-α) production in human peripheral blood mononuclear cells (PBMCs) in a sequence-dependent manner, indicative of TLR7/8 recruitment. The effect of inosine incorporation on TLR7/8 sensing was restricted to immunostimulatory ssRNAs and was not seen with inosine-containing short double-stranded RNAs or with a deoxy-inosine-modified ssRNA. Inosine-mediated increase of self-secondary structure of an ssRNA resulted in potentiated IFN-α production in human PBMCs through TLR7 recruitment, as established through the use of a TLR7 antagonist and Tlr7-deficient cells. There was a correlation between hyperediting of influenza A viral ssRNA and its ability to stimulate TNF-α, independent of 5'-triphosphate residues, and involving Adar-1. Furthermore, A-to-I editing of viral ssRNA directly enhanced mouse Tlr7 sensing, when present in proportions reproducing biologically relevant levels of RNA editing. Thus, we demonstrate for the first time that inosine incorporation into immunostimulatory ssRNA can potentiate TLR7/8 activation. Our results suggest a novel function of A-to-I RNA editing, which is to facilitate TLR7/8 sensing of phagocytosed viral RNA.


Subject(s)
Influenza A Virus, H1N1 Subtype/genetics , Influenza, Human/immunology , Influenza, Human/virology , Inosine/genetics , RNA Editing , RNA, Viral/genetics , Toll-Like Receptor 7/immunology , Toll-Like Receptor 8/immunology , Adenosine/genetics , Adenosine/immunology , Adenosine Deaminase/genetics , Adenosine Deaminase/immunology , Animals , Base Sequence , Female , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/genetics , Inosine/immunology , Interferon-alpha/genetics , Interferon-alpha/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Male , Mice , Mice, Knockout , Molecular Sequence Data , Nucleic Acid Conformation , RNA, Viral/chemistry , RNA, Viral/immunology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 8/genetics
SELECTION OF CITATIONS
SEARCH DETAIL