Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Immunol Cell Biol ; 102(2): 117-130, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38069638

ABSTRACT

Programmed death receptor 1 (PD-1) is an inhibitory receptor on T cells shown to restrain T-cell proliferation. PD-1 immune checkpoint blockade has emerged as a highly promising approach in cancer treatment. Much of our understanding of the function of PD-1 is derived from in vitro T-cell activation assays. Here we set out to further investigate how T cells integrate inhibitory signals such as PD-1 in vitro using the PD-1 agonist, PD-1 ligand 1 (PD-L1) fusion protein (PD-L1.Fc), coimmobilized alongside anti-CD3 agonist monoclonal antibody (mAb) on plates to deliver PD-1 signals to wild-type and PD-1-/- CD8+ T cells. Surprisingly, we found that the PD-L1.Fc fusion protein inhibited T-cell proliferation independently of PD-1. This PD-L1.Fc inhibition was observed in the presence and absence of CD28 and interleukin-2 signaling. Binding of PD-L1.Fc was restricted to PD-1-expressing T cells and thus inhibition was not mediated by the interaction of PD-L1.Fc with CD80 or other yet unknown binding partners. Furthermore, a similar PD-1-independent reduction of T-cell proliferation was observed with plate-bound PD-L2.Fc. Hence, our results suggest that the coimmobilization of PD-1 ligand fusion proteins with anti-CD3 mAb leads to a reduction of T-cell engagement with plate-bound anti-CD3 mAb. This study demonstrates a nonspecific mechanism of T-cell inhibition when PD-L1.Fc or PD-L2.Fc fusion proteins are delivered in a plate-bound coimmobilization assay and highlights the importance of careful optimization of assay systems and reagents when interpreting their influence on T-cell proliferation.


Subject(s)
CD8-Positive T-Lymphocytes , Programmed Cell Death 1 Receptor , Programmed Cell Death 1 Receptor/metabolism , B7-H1 Antigen/metabolism , Ligands , Cell Proliferation , Receptors, Death Domain/metabolism
2.
Immunity ; 41(1): 104-15, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25035955

ABSTRACT

The relationship between dendritic cells (DCs) and macrophages is often debated. Here we ask whether steady-state, lymphoid-tissue-resident conventional DCs (cDCs), plasmacytoid DCs (pDCs), and macrophages share a common macrophage-DC-restricted precursor (MDP). Using new clonal culture assays combined with adoptive transfer, we found that MDP fractions isolated by previous strategies are dominated by precursors of macrophages and monocytes, include some multipotent precursors of other hematopoietic lineages, but contain few precursors of resident cDCs and pDCs and no detectable common precursors restricted to these DC types and macrophages. Overall we find no evidence for a common restricted MDP leading to both macrophages and FL-dependent, resident cDCs and pDCs.


Subject(s)
Cell Lineage/immunology , Dendritic Cells/cytology , Lymphoid Tissue/cytology , Macrophages/cytology , Monocyte-Macrophage Precursor Cells/cytology , Adoptive Transfer , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , CX3C Chemokine Receptor 1 , Cell Differentiation/immunology , Cells, Cultured , Cytokines/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocytes/cytology , Granulocytes/immunology , Macrophage Colony-Stimulating Factor/immunology , Mice , Mice, Inbred C57BL , Monocyte-Macrophage Precursor Cells/immunology , Monocytes/cytology , Receptor, Macrophage Colony-Stimulating Factor/immunology , Receptors, Chemokine/immunology
3.
J Immunol ; 205(7): 1842-1856, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32839238

ABSTRACT

Follicular dendritic cells and macrophages have been strongly implicated in presentation of native Ag to B cells. This property has also occasionally been attributed to conventional dendritic cells (cDC) but is generally masked by their essential role in T cell priming. cDC can be divided into two main subsets, cDC1 and cDC2, with recent evidence suggesting that cDC2 are primarily responsible for initiating B cell and T follicular helper responses. This conclusion is, however, at odds with evidence that targeting Ag to Clec9A (DNGR1), expressed by cDC1, induces strong humoral responses. In this study, we reveal that murine cDC1 interact extensively with B cells at the border of B cell follicles and, when Ag is targeted to Clec9A, can display native Ag for B cell activation. This leads to efficient induction of humoral immunity. Our findings indicate that surface display of native Ag on cDC with access to both T and B cells is key to efficient humoral vaccination.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Lectins, C-Type/metabolism , Receptors, Immunologic/metabolism , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Antigen Presentation , Autoantigens/immunology , Autoantigens/metabolism , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Immunity, Humoral , Lectins, C-Type/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Immunologic/genetics , Vaccination
4.
Proc Natl Acad Sci U S A ; 112(13): 4044-9, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25775525

ABSTRACT

Dendritic cells (DCs) are heterogeneous, comprising subsets with functional specializations that play distinct roles in immunity as well as immunopathology. We investigated the molecular control of cell survival of two main DC subsets: plasmacytoid DCs (pDCs) and conventional DCs (cDCs) and their dependence on individual antiapoptotic BCL-2 family members. Compared with cDCs, pDCs had higher expression of BCL-2, lower A1, and similar levels of MCL-1 and BCL-XL. Transgenic overexpression of BCL-2 increased the pDC pool size in vivo with only minor impact on cDCs. With a view to immune intervention, we tested BCL-2 inhibitors and found that ABT-199 (the BCL-2 specific inhibitor) selectively killed pDCs but not cDCs. Conversely, genetic knockdown of A1 profoundly reduced the proportion of cDCs but not pDCs. We also found that conditional ablation of MCL-1 significantly reduced the size of both DC populations in mice and impeded DC-mediated immune responses. Thus, we revealed that the two DC types have different cell survival requirements. The molecular basis of survival of different DC subsets thus advocates the antagonism of selective BCL-2 family members for treating diseases pertaining to distinct DC subsets.


Subject(s)
Apoptosis , Dendritic Cells/cytology , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Cell Separation , Cell Survival , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Signal Transduction , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/cytology , Transgenes , bcl-X Protein/metabolism
5.
J Immunol ; 192(5): 2202-9, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24489100

ABSTRACT

Although multiple dendritic cell (DC) subsets have the potential to induce Th17 differentiation in vitro, the key DC that is critical in Th17 induction and Th17-mediated disease remains moot. In this study, we revealed that CCR2(+) monocyte-derived DCs (moDCs), but not conventional DCs, were critical for in vivo Th17 induction and autoimmune inflammation. Functional comparison in vitro indicated that moDCs are the most potent type of Th17-inducing DCs compared with conventional DCs and plasmacytoid DCs. Furthermore, we demonstrated that the importance of GM-CSF in Th17 induction and Th17-mediated disease is its endowment of moDCs to induce Th17 differentiation in vivo, although it has little effect on moDC numbers. Our findings identify the in vivo cellular targets that can be selectively manipulated to ameliorate Th17-mediated inflammatory diseases, as well as the mechanism of GM-CSF antagonism in such diseases.


Subject(s)
Autoimmune Diseases/immunology , Cell Differentiation/immunology , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Monocytes/immunology , Th17 Cells/immunology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Cell Differentiation/genetics , Dendritic Cells/cytology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Mice , Mice, Knockout , Monocytes/cytology , Th17 Cells/cytology
6.
Blood ; 121(1): 11-9, 2013 Jan 03.
Article in English | MEDLINE | ID: mdl-23053574

ABSTRACT

The developmental origin of IFN-producing plasmacytoid dendritic cells (pDCs) has been uncertain. In the present study, we tracked the development of pDCs in cultures of BM precursors stimulated with Flt3 ligand. Common myeloid precursors (CMPs) produced both conventional DCs (cDCs) and pDCs via the DC-restricted common DC precursor. Common lymphoid precursors (CLPs) produced only a few cDCs with variable efficiency, but produced pDCs via a transient intermediate precursor with B-cell potential. The pDCs of both origins produced IFN-α when stimulated with CpG oligonucleotides. The pDCs of CLP origin showed evidence of past RAG1 expression and had D-J rearrangements in IgH genes. Most pDCs and all cDCs of CMP origin lacked these signs of a lymphoid past. However, in these cultures, some pDCs of CMP origin showed evidence of past RAG1 expression and had D-J IgH gene rearrangements; most of these derived from a subset of CMPs already expressing RAG1.


Subject(s)
Dendritic Cells/cytology , Lymphopoiesis/physiology , Myelopoiesis/physiology , Adoptive Transfer , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Bone Marrow Cells/classification , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Lineage , Cell Separation , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Clone Cells/cytology , Clone Cells/metabolism , CpG Islands , Dendritic Cells/metabolism , Gene Expression Regulation, Developmental , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Immunophenotyping , Interferon-alpha/biosynthesis , Interferon-alpha/genetics , Lymphopoiesis/genetics , Membrane Proteins/pharmacology , Mice , Mice, Inbred C57BL , Myelopoiesis/genetics , Oligonucleotides/pharmacology , Radiation Chimera , Specific Pathogen-Free Organisms
7.
Cell Death Differ ; 31(2): 150-158, 2024 02.
Article in English | MEDLINE | ID: mdl-38097622

ABSTRACT

Whole-genome screens using CRISPR technologies are powerful tools to identify novel tumour suppressors as well as factors that impact responses of malignant cells to anti-cancer agents. Applying this methodology to lymphoma cells, we conducted a genome-wide screen to identify novel inhibitors of tumour expansion that are induced by the tumour suppressor TRP53. We discovered that the absence of Arrestin domain containing 3 (ARRDC3) increases the survival and long-term competitiveness of MYC-driven lymphoma cells when treated with anti-cancer agents that activate TRP53. Deleting Arrdc3 in mice caused perinatal lethality due to various developmental abnormalities, including cardiac defects. Notably, the absence of ARRDC3 markedly accelerated MYC-driven lymphoma development. Thus, ARRDC3 is a new mediator of TRP53-mediated suppression of tumour expansion, and this discovery may open new avenues to harness this process for cancer therapy.


Subject(s)
Lymphoma , Neoplasms , Animals , Mice , Arrestins/genetics , Arrestins/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Neoplasms/genetics
8.
J Immunol ; 186(9): 5184-92, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21422244

ABSTRACT

The development of Ag-presenting functions by murine dendritic cells (DCs) of the CD8(+) DC lineage was studied using a Flt-3 ligand stimulated bone-marrow culture system. Although newly formed DCs of this lineage are capable of Ag uptake and efficient presentation to T cells on MHC class II, they initially lack the ability to cross-present exogenous Ags on MHC class I. Cross-presentation capacity is acquired as a subsequent maturation step, promoted by cytokines such as GM-CSF. The development of cross-presentation capacity by the DCs in these cultures may be monitored by the parallel development of DC surface expression of CD103. However, the expression of CD103 and cross-presentation capacity are not always linked; therefore, CD103 is not an essential part of the cross-presentation machinery. These results explain the considerable variability in CD103 expression by CD8(+) DCs as well as the findings that not all DCs of this lineage are capable of cross-presentation.


Subject(s)
Antigen Presentation/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Animals , Antigens, CD/biosynthesis , Antigens, CD/immunology , CD8 Antigens/biosynthesis , CD8 Antigens/immunology , Cell Differentiation/immunology , Cell Separation , Dendritic Cells/cytology , Dendritic Cells/metabolism , Flow Cytometry , Integrin alpha Chains/biosynthesis , Integrin alpha Chains/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic
9.
J Immunol ; 186(11): 6207-17, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21515786

ABSTRACT

Mouse dendritic cells (DC) have been extensively studied in various tissues, especially spleen, and they comprise subsets with distinct developmental origins, surface phenotypes, and functions. Considerably less is known about human DC due to their rarity in blood and inaccessibility of other human tissues. The study of DC in human blood has revealed four subsets distinct in phenotype and function. In this study, we describe four equivalent DC subsets in human spleen obtained from deceased organ donors. We identify three conventional DC subsets characterized by surface expression of CD1b/c, CD141, and CD16, and one plasmacytoid DC subset characterized by CD304 expression. Human DC subsets in spleen were very similar to those in human blood with respect to surface phenotype, TLR and transcription factor expression, capacity to stimulate T cells, cytokine secretion, and cross-presentation of exogenous Ag. However, organ donor health status, in particular treatment with corticosteroid methylprednisolone and brain death, may affect DC phenotype and function. DC T cell stimulatory capacity was reduced but DC were qualitatively unchanged in methylprednisolone-treated deceased organ donor spleen compared with healthy donor blood. Overall, our findings indicate that human blood DC closely resemble human spleen DC. Furthermore, we confirm parallels between human and mouse DC subsets in phenotype and function, but also identify differences in transcription factor and TLR expression as well as functional properties. In particular, the hallmark functions of mouse CD8α(+) DC subsets, that is, IL-12p70 secretion and cross-presentation, are not confined to the equivalent human CD141(+) DC but are shared by CD1b/c(+) and CD16(+) DC subsets.


Subject(s)
Cross-Priming/immunology , Dendritic Cells/immunology , Spleen/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Aged , Animals , CD11c Antigen/immunology , CD11c Antigen/metabolism , Cells, Cultured , Dendritic Cells/metabolism , Female , Flow Cytometry , HLA-DR Antigens/immunology , HLA-DR Antigens/metabolism , Health Status , Heart Diseases/blood , Heart Diseases/immunology , Heart Diseases/pathology , Humans , Hypertension/blood , Hypertension/immunology , Hypertension/pathology , Immunophenotyping , Interleukin-12/immunology , Interleukin-12/metabolism , Male , Mice , Middle Aged , Receptors, IgG/immunology , Receptors, IgG/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spleen/cytology , Spleen/metabolism , T-Lymphocytes/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism
10.
J Immunol ; 187(2): 842-50, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21677141

ABSTRACT

Three surface molecules of mouse CD8(+) dendritic cells (DCs), also found on the equivalent human DC subpopulation, were compared as targets for Ab-mediated delivery of Ags, a developing strategy for vaccination. For the production of cytotoxic T cells, DEC-205 and Clec9A, but not Clec12A, were effective targets, although only in the presence of adjuvants. For Ab production, however, Clec9A excelled as a target, even in the absence of adjuvant. Potent humoral immunity was a result of the highly specific expression of Clec9A on DCs, which allowed longer residence of targeting Abs in the bloodstream, prolonged DC Ag presentation, and extended CD4 T cell proliferation, all of which drove highly efficient development of follicular helper T cells. Because Clec9A shows a similar expression pattern on human DCs, it has particular promise as a target for vaccines of human application.


Subject(s)
Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , Cytotoxicity Tests, Immunologic , Dendritic Cells/immunology , Immunophenotyping , Lectins, C-Type/metabolism , Receptors, Immunologic/metabolism , Adjuvants, Immunologic/administration & dosage , Animals , Antigen Presentation/genetics , Antigens, CD/genetics , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/classification , CD4-Positive T-Lymphocytes/metabolism , Cytotoxicity Tests, Immunologic/methods , Dendritic Cells/metabolism , Humans , Immunophenotyping/methods , Lectins, C-Type/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Minor Histocompatibility Antigens , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Immunologic/genetics , Receptors, Mitogen/genetics , Receptors, Mitogen/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Vaccines, DNA/chemical synthesis , Vaccines, DNA/genetics , Vaccines, DNA/immunology
11.
J Exp Med ; 196(10): 1307-19, 2002 Nov 18.
Article in English | MEDLINE | ID: mdl-12438422

ABSTRACT

The CD45RA(hi)CD11c(int) plasmacytoid predendritic cells (p-preDCs) of mouse lymphoid organs, when stimulated in culture with CpG or influenza virus, produce large amounts of type I interferons and transform without division into CD8(+)CD205(-) DCs. P-preDCs express CIRE, the murine equivalent of DC-specific intercellular adhesion molecule 3 grabbing nonintegrin (DC-SIGN). P-preDCs are divisible by CD4 expression into two subgroups differing in turnover rate and in response to Staphylococcus aureus. The kinetics of bromodeoxyuridine labeling and the results of transfer to normal recipient mice indicate that CD4(-) p-preDCs are the immediate precursors of CD4(+) p-preDCs. Similar experiments indicate that p-preDCs are normally long lived and are not the precursors of the short-lived steady-state conventional DCs. However, in line with the culture studies on transfer to influenza virus-stimulated mice the p-preDCs transform into CD8(+)CD205(-) DCs, distinct from conventional CD8(+)CD205(+) DCs. Hence as well as activating preexistant DCs, microbial infection induces a wave of production of a new DC subtype. The functional implications of this shift in the DC network remain to be determined.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Dendritic Cells/cytology , Orthomyxoviridae/immunology , Staphylococcus aureus/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Female , Lymphoid Tissue/cytology , Male , Mice , Mice, Inbred C57BL , Phenotype
12.
Blood ; 112(8): 3264-73, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18669894

ABSTRACT

A novel dendritic cell (DC)-restricted molecule, Clec9A, was identified by gene expression profiling of mouse DC subtypes. Based on sequence similarity, a human ortholog was identified. Clec9A encodes a type II membrane protein with a single extracellular C-type lectin domain. Both the mouse Clec9A and human CLEC9A were cloned and expressed, and monoclonal antibodies (mAbs) against each were generated. Surface staining revealed that Clec9A was selective for mouse DCs and was restricted to the CD8(+) conventional DC and plasmacytoid DC subtypes. A subset of human blood DCs also expressed CLEC9A. A single injection of mice with a mAb against Clec9A, which targets antigens (Ags) to the DCs, produced a striking enhancement of antibody responses in the absence of added adjuvants or danger signals, even in mice lacking Toll-like receptor signaling pathways. Such targeting also enhanced CD4 and CD8 T-cell responses. Thus, Clec9A serves as a new marker to distinguish subtypes of both mouse and human DCs. Furthermore, targeting Ags to DCs with antibodies to Clec9A is a promising strategy to enhance the efficiency of vaccines, even in the absence of adjuvants.


Subject(s)
Dendritic Cells/cytology , Lectins, C-Type/chemistry , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Hematopoietic Stem Cells/cytology , Humans , Lectins, C-Type/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Signal Transduction , Vaccines/chemistry , Vaccines/metabolism
13.
Methods Mol Biol ; 415: 163-78, 2008.
Article in English | MEDLINE | ID: mdl-18370154

ABSTRACT

Dendritic cells (DC) are widely regarded as the most potent cellular inducers of the adaptive immune response; so, immunotherapy through DC manipulation is a promising option in the future fight against many human ailments. We have developed a method of isolating DC from the mouse that involves efficient extraction from tissues, followed by the selection of the lightest density cells, then depletion of non-DC through a cocktail of monoclonal antibodies and anti-immunoglobulin magnetic beads. Finally, purification and segregation into DC subtypes is achieved by immunofluorescent labeling and sorting. This has demonstrated a network of DC populations differing in surface phenotype and function. We can now produce larger numbers of many of these DC subpopulations from their precursors using bone marrow cultures supplemented with fms-like tyrosine kinase 3 ligand (Flt3L). The culture-generated DC can be aligned with the populations directly isolated from tissues. Combining the in vivo and in vitro systems will make study of murine DC and their alignment to their human counterparts an easier break process.


Subject(s)
Cell Culture Techniques/methods , Cell Separation/methods , Dendritic Cells/cytology , Lymphoid Tissue/cytology , Animals , Antibodies, Monoclonal , Antigens, Surface/metabolism , Bone Marrow , Cell Lineage , Flow Cytometry , Fluorescent Antibody Technique , Immunomagnetic Separation , Membrane Proteins/metabolism , Mice
14.
Methods Mol Biol ; 1423: 61-87, 2016.
Article in English | MEDLINE | ID: mdl-27142009

ABSTRACT

Dendritic cells (DCs) form a complex network of cells that initiate and orchestrate immune responses against a vast array of pathogenic challenges. Developmentally and functionally distinct DC subtypes differentially regulate T-cell function. Importantly it is the ability of DC to capture and process antigen, whether from pathogens, vaccines, or self-components, and present it to naive T cells that is the key to their ability to initiate an immune response. Our typical isolation procedure for DC from murine spleen was designed to efficiently extract all DC subtypes, without bias and without alteration to their in vivo phenotype, and involves a short collagenase digestion of the tissue, followed by selection for cells of light density and finally negative selection for DC. The isolation procedure can accommodate DC numbers that have been artificially increased via administration of fms-like tyrosine kinase 3 ligand (Flt3L), either directly through a series of subcutaneous injections or by seeding with an Flt3L secreting murine melanoma. Flt3L may also be added to bone marrow cultures to produce large numbers of in vitro equivalents of the spleen DC subsets. Total DC, or their subsets, may be further purified using immunofluorescent labeling and flow cytometric cell sorting. Cell sorting may be completely bypassed by separating DC subsets using a combination of fluorescent antibody labeling and anti-fluorochrome magnetic beads. Our procedure enables efficient separation of the distinct DC subsets, even in cases where mouse numbers or flow cytometric cell sorting time is limiting.


Subject(s)
Dendritic Cells/cytology , Immunomagnetic Separation/methods , Spleen/cytology , Animals , Bone Marrow Cells/cytology , Cell Separation/methods , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/immunology , Flow Cytometry/methods , Membrane Proteins/administration & dosage , Membrane Proteins/pharmacology , Mice , Spleen/drug effects
15.
Mol Immunol ; 63(2): 264-7, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25081090

ABSTRACT

When mouse dendritic cells (DCs) are isolated from tissues, purified and placed in a nutritive culture they die more rapidly than would be expected from their normal turnover in vivo. This can distort culture assays of DC function. We therefore tested several approaches to prolonging DC survival in culture. Of several cytokines tested granulocyte-macrophage colony stimulating factor was most effective at preserving the viability of conventional DCs (cDCs) but was ineffective for plasmacytoid DCs (pDCs). Surprisingly, Fms-like tyrosine kinase 3 ligand, crucial for DC development, produced only a marginal improvement in DC survival in culture, and interleukin-3, reported to prevent apoptosis of human pDCs, produced only a minor improvement in survival of mouse DCs. Genetic manipulation of cell death pathways was also tested, to avoid activation effects exerted by cytokine signalling. The isolation of DCs from mice overexpressing Bcl-2 was especially effective in maintaining pDC viability but gave a lesser improvement in cDC viability. DCs isolated from Bim(-/-)Noxa(-/-) mice also showed improved culture survival, but in this case with pDCs showing the least improvement.


Subject(s)
Cell Culture Techniques/methods , Dendritic Cells/cytology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Survival/drug effects , Cells, Cultured , Cytokines/pharmacology , Dendritic Cells/drug effects , Humans , Mice, Inbred C57BL , Signal Transduction/drug effects , Signal Transduction/genetics
16.
Arthritis Rheumatol ; 67(3): 797-808, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25418983

ABSTRACT

OBJECTIVE: Interferon-α (IFNα)-producing plasmacytoid dendritic cells (PDCs) are implicated in the pathogenesis of systemic lupus erythematosus (SLE). IFNα-related genes are highlighted among SLE susceptibility alleles and are characteristically expressed in the blood of patients with SLE, while in mouse models of lupus, PDC numbers and IFNα production are increased. This study was undertaken to investigate the effects of inhibitors that selectively target different antiapoptotic molecules on the survival of PDCs. METHODS: PDC numbers, in vitro survival, and expression of antiapoptotic molecules were evaluated in lupus-prone (NZB × NZW)F1 (NZB/NZW) mice. The impact of Bcl-2 antagonists and glucocorticoids on PDCs was evaluated in vitro and in vivo. IFNα production by NZB/NZW mice was evaluated before and after treatment with Bcl-2 antagonists. RESULTS: PDCs, but not lymphoid tissue-resident conventional DCs, largely relied on the antiapoptotic protein Bcl-2 for survival. The enlarged PDC compartment in NZB/NZW mice was associated with selectively prolonged survival and increased Bcl-2 transcription. Functionally, this resulted in enhanced production of IFNα. Bcl-2 inhibitors selectively killed mouse and human PDCs, including PDCs from SLE patients, but not conventional DCs, dampened IFNα production by PDCs, and synergized with glucocorticoids to kill activated PDCs. CONCLUSION: Enhanced PDC survival is a likely contributing factor to enhanced IFNα production by lupus PDCs. Bcl-2 antagonists potently and selectively kill PDCs and reduce IFNα production. Thus, we believe that they are attractive candidates for treating PDC-associated diseases.


Subject(s)
Biphenyl Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Dendritic Cells/drug effects , Interferon-alpha/metabolism , Lupus Erythematosus, Systemic/immunology , Nitrophenols/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Annexin A5/metabolism , Antibodies, Antinuclear/blood , Cell Survival , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/pathology , Flow Cytometry , Glucocorticoids/pharmacology , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NZB , Mice, Transgenic , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Real-Time Polymerase Chain Reaction
17.
Methods Mol Biol ; 960: 327-350, 2013.
Article in English | MEDLINE | ID: mdl-23329497

ABSTRACT

Dendritic cells (DC) are found at low frequency in lymphoid and non-lymphoid tissues. Different DC subsets are adept at different roles in immunity in diverse scenarios of attack by infectious agents, as well as in the maintenance of self-tolerance. A key element in the ability of DC to initiate adaptive immune responses is their capacity to capture and process antigen, whether from pathogens, vaccines or self-components, and present it to T cells. Our typical procedure for isolation of the different DC types from murine spleen involves their digestion from the tissue using collagenase, selection of cells of light density, and negative selection for DC. DC may then be separated into their functionally distinct subpopulations using immunofluorescent labeling and flow cytometric cell sorting. If the availability of mice is limiting, our protocol can cater for DC numbers boosted by the administration of fms-like tyrosine kinase 3 ligand (Flt3L), directly via subcutaneous injection or via the introduction of a Flt3L secreting melanoma cell line. Large numbers of in vitro equivalents of the spleen DC subsets may also be produced by culturing bone marrow with Flt3L. If flow cytometric sorting time is a limitation splenic DC subpopulations may instead be separated using a combination of fluorescent antibody labeling and anti-fluorochrome magnetic beads. Careful segregation of these functionally distinct subpopulations of DC will enable a thorough examination of their antigen processing and presenting capabilities.


Subject(s)
Cell Separation/methods , Dendritic Cells/cytology , Spleen/immunology , Animals , Cell Count , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Deoxyribonuclease I/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Immunomagnetic Separation , Ligands , Mice , Tissue and Organ Harvesting , fms-Like Tyrosine Kinase 3/metabolism
18.
Adv Immunol ; 120: 105-26, 2013.
Article in English | MEDLINE | ID: mdl-24070382

ABSTRACT

Interferon-producing plasmacytoid dendritic cells (pDC) are a specialized branch of the dendritic cell (DC) family, and their differentiation in mice is closely linked to that of conventional DC (cDC). Several different developmental pathways retain the potential to form pDC and are likely to contribute to the steady-state pDC population. A lymphoid pathway to DC development produces mainly pDC as a branch otherwise leading to B-cell development; such pDC may carry relics of a lymphoid past such as DJ rearrangements of immunoglobulin heavy chain (IgH) genes. The myeloid pathway to pDC and cDC is better known, but recent reassessment has revealed several substreams of development with separate DC-committed precursors. One substream has a lymphoid-like aspect, involving a precursor expressing RAG-1 and producing pDC with IgH gene rearrangements. Another more biased to cDC production produces pDC without such IgH gene rearrangements. Finally, there is the production of interferon-producing pDC-like cells that are not pDC but appear to be cDC precursors; these do not express key pDC markers such as CCR9. Initiation of the DC and then the pDC developmental program overrides any surface marker-expressed developmental bias to other myeloid or lymphoid lineages, resulting in an apparent convergent differentiation to the pDC form. A DC fate is sometimes imprinted early in development, upstream of identifiable myeloid, or lymphoid precursors. This suggests that DC, including pDC, represent a distinct hematopoietic lineage separate from conventional myeloid or lymphoid cells.


Subject(s)
Dendritic Cells/cytology , Animals , Cell Lineage , Dendritic Cells/metabolism , Hematopoiesis , Humans
19.
Mol Immunol ; 50(1-2): 9-17, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22209163

ABSTRACT

Injection of antigens coupled to antibodies against the dendritic cell (DC) surface molecule Clec9A has been shown to produce strongly enhanced antibody responses even without co-administration of adjuvants, via antigen presentation by DC on MHC class II and consequent production of follicular helper T cells. A series of mutant mice were tested to determine the DC subtypes responsible for this MHC II presentation of targeted antigen, compared to presentation of antigen on MHC I. A new clec9A null mouse was developed; these mice did not give enhanced antibody production, confirming the response was dependent on Clec9A-expressing DC. However targeting of antigen to Clec9A in batf3 null mice produced enhanced antibody responses despite the marked reduction in CD8(+) DC, the major Clec9A-expressing DC subtype. This was shown to be dependent on efficient MHC II presentation by minor Clec9A-expressing DC subtypes in the environment of the Batf3(-/-) mice, namely early cells of the CD8 DC lineage and the plasmacytoid-related CD8(+) DC subset, but not by plasmacytoid cells themselves. However in normal mice most MHC II presentation of the Clec9A-targeted antigen was by the major CD8(+) DC population, the DC also responsible for presentation on MHC I.


Subject(s)
Antibody Formation/immunology , Basic-Leucine Zipper Transcription Factors/immunology , Dendritic Cells/immunology , Lectins, C-Type/immunology , Receptors, Immunologic/immunology , Repressor Proteins/immunology , Animals , Antigen Presentation/immunology , Basic-Leucine Zipper Transcription Factors/genetics , CD8 Antigens/immunology , Cells, Cultured , Dendritic Cells/metabolism , Female , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Lectins, C-Type/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/genetics , Ovalbumin/immunology , Receptors, Immunologic/genetics , Repressor Proteins/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
20.
Innate Immun ; 17(3): 338-52, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20501515

ABSTRACT

Dendritic cells (DCs) serve as a link between the innate and adaptive immune systems. The activation state of DCs is crucial in this role. However, when DCs are isolated from lymphoid tissues, purified and placed in culture they undergo 'spontaneous' activation. The basis of this was explored, using up-regulation of DC surface MHC II, CD40, CD80 and CD86 as indicators of DC activation. No evidence was found for DC damage during isolation or for microbial products causing the activation. The culture activation of spleen DCs differed from that of Langerhans cells when released from E-cadherin-mediated adhesions, since E-cadherin was not detected and activation still occurred with ß-catenin null DCs. Much of the activation could be attributed to DC-DC interactions. Although increases in surface MHC II levels occurred under all culture conditions tested, the increase in expression of CD40, CD80 and CD86 was much less under culture conditions where such interactions were minimised. DC-to-DC contact under the artificial conditions of high DC concentration in culture induced the production of soluble factors and these, in turn, induced the up-regulation of co-stimulatory molecules on the DC surface.


Subject(s)
Antigens, CD/metabolism , Cell Communication , Cytokines/metabolism , Dendritic Cells/metabolism , Histocompatibility Antigens Class II/metabolism , Animals , Antigens, CD/genetics , Cadherins/metabolism , Cell Adhesion , Cell Communication/immunology , Cell Differentiation , Cells, Cultured , Chimera , Cytokines/genetics , Dendritic Cells/immunology , Dendritic Cells/pathology , Histocompatibility Antigens Class II/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Spleen , Up-Regulation/immunology , beta Catenin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL