Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Nat Immunol ; 14(6): 584-92, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23624556

ABSTRACT

Interleukin 17 (IL-17)-committed γδ T cells (γδT17 cells) participate in many immune responses, but their developmental requirements and subset specific functions remain poorly understood. Here we report that a commonly used CD45.1(+) congenic C57BL/6 mouse substrain is characterized by selective deficiency in Vγ4(+) γδT17 cells. This trait was due to a spontaneous mutation in the gene encoding the transcription factor Sox13 that caused an intrinsic defect in development of those cells in the neonatal thymus. The γδT17 cells migrated from skin to lymph nodes at low rates. In a model of psoriasis-like dermatitis, the Vγ4(+) γδT17 cell subset expanded considerably in lymph nodes and homed to inflamed skin. Sox13-mutant mice were protected from psoriasis-like skin changes, which identified a role for Sox13-dependent γδT17 cells in this inflammatory condition.


Subject(s)
Autoantigens/immunology , Dermatitis/immunology , Interleukin-17/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Animals , Animals, Newborn , Autoantigens/genetics , Autoantigens/metabolism , Cells, Cultured , Dermatitis/genetics , Dermatitis/metabolism , Flow Cytometry , Interleukin-17/genetics , Interleukin-17/metabolism , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Leukocyte Common Antigens/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Inbred NOD , Mice, Knockout , Mutation , Psoriasis/genetics , Psoriasis/immunology , Psoriasis/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Thymus Gland/pathology
2.
Immunity ; 43(2): 382-93, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26287683

ABSTRACT

Macrophages are one of the most diverse cell populations in terms of their anatomical location and functional specialization during both homeostasis and disease. Although it has been shown in different fate mapping models that some macrophages present in adult tissues are already established during fetal development, their exact origins are still under debate. In the current study, we developed a fate mapping strain, based on the Kit locus, which allowed us to readdress "the origins" question. Different types of macrophages from various adult tissues were traced to their fetal or adult sources by inducing labeling in precursors at several time points either during fetal development or in adult mice. We show that all adult macrophages, resident or infiltrating, are progenies of classical hematopoietic stem cells (HSC) with the exception of microglia and, partially epidermal Langerhans cells, which are yolk sac (YS)-derived.


Subject(s)
Fetal Development/immunology , Hematopoietic Stem Cells/physiology , Macrophages/physiology , Microglia/physiology , Proto-Oncogene Proteins c-kit/metabolism , Animals , Antigens, CD/metabolism , Cell Differentiation , Cell Lineage , Embryo, Mammalian , Female , Homeostasis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Proto-Oncogene Proteins c-kit/genetics , Yolk Sac/physiology
4.
J Immunol ; 199(10): 3691-3699, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29030488

ABSTRACT

Intestinal IL-17-producing cells, including Th17, γ/δ T, and innate lymphoid cells, are differentially distributed along the gastrointestinal tract. In this study, we show that the gut IL-17-producing γ/δ T (γ/δ T17) cells develop before birth and persist in the tissue as long-lived cells with minimal turnover. Most colon γ/δ T17 cells express, together with Vγ4 and CCR6, the scavenger receptor 2 and are mainly restricted to innate lymphoid follicles in the colon. Colon γ/δ T cells in mice that lack conventional dendritic cells 2 produced increased amounts of IL-17 with concomitant heightened epithelial antimicrobial response, such as the C-type lectins Reg3γ and Reg3ß. In the absence of γ/δ T cells or after IL-17 neutralization, this epithelial response was dramatically reduced, underlining the protective role of this unique subpopulation of innate γ/δ T17 cells in the colonic mucosa.


Subject(s)
Anti-Infective Agents/metabolism , Colon/immunology , Epithelial Cells/immunology , Intestinal Mucosa/immunology , Pancreatitis-Associated Proteins/metabolism , T-Lymphocytes/immunology , Animals , Cell Differentiation , Cells, Cultured , Fetal Development , Immunity, Innate , Interleukin-17/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, CCR6/metabolism , Receptors, Scavenger/metabolism
5.
Eur J Immunol ; 44(7): 2003-12, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24687623

ABSTRACT

Alveolar macrophages (AMs), localized at the pulmonary air-tissue interface, are one of the first lines of defense that interact with inhaled airborne pathogens such as influenza viruses. By using a new CD169-DTR transgenic mouse strain we demonstrate that specific and highly controlled in vivo ablation of this myeloid cell subset leads to severe impairment of the innate, but not adaptive, immune responses and critically affects the progression of the disease. In fact, AM-ablated mice, infected with a normally sublethal dose of PR8 influenza virus, showed dramatically increased virus load in the lungs, severe airway inflammation, pulmonary edema and vascular leakage, which caused the death of the infected animals. Our data highlight the possibilities for new therapeutic strategies focusing on modulation of AMs, which may efficiently boost innate responses to influenza infections.


Subject(s)
Adaptive Immunity , Influenza A Virus, H1N1 Subtype , Macrophages, Alveolar/physiology , Orthomyxoviridae Infections/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/physiology , Female , Lung/virology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Sialic Acid Binding Ig-like Lectin 1/physiology , Viral Load
6.
Eur J Immunol ; 44(4): 1181-93, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24374622

ABSTRACT

Cellular differentiation of the T-cell branch of the immune system begins with the HSC, which undergoes a series of stages characterized by progressive restriction in multipotency and acquisition of specific lineage identity At the molecular level, the restriction of cell potential, commitment, and differentiation to a specific lineage is achieved through the coordinated control of gene expression and epigenetic mechanisms. Here, we analyzed and compared the gene expression profiles and the genome-wide histone modification marks H3K4me3 (H3 lysine 4 trimethylation) and H3K27me3 (H3 lysine 27 trimethylation) in (i) in vitro propagated HSCs, (ii) in vitro generated and propagated pro-T cells derived from these stem cells, and (iii) double-positive thymocytes derived from these pro-T cells after injection into Rag-deficient mice. The combined analyses of the different datasets in this unique experimental system highlighted the importance of both transcriptional and epigenetic repression in shaping the early phases of T-cell development.


Subject(s)
Epigenesis, Genetic , Epigenomics/methods , Multipotent Stem Cells/metabolism , T-Lymphocytes/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , Cluster Analysis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Histones/metabolism , Lysine/metabolism , Methylation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Multipotent Stem Cells/cytology , Oligonucleotide Array Sequence Analysis , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/metabolism , T-Lymphocytes/cytology
7.
J Immunol ; 189(3): 1128-32, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22732587

ABSTRACT

Plasmodium infections trigger strong innate and acquired immune responses, which can lead to severe complications, including the most feared and often fatal cerebral malaria (CM). To begin to dissect the roles of different dendritic cell (DC) subsets in Plasmodium-induced pathology, we have generated a transgenic strain, Clec9A-diphtheria toxin receptor that allows us to ablate in vivo Clec9A(+) DCs. Specifically, we have analyzed the in vivo contribution of this DC subset in an experimental CM model using Plasmodium berghei, and we provide strong evidence that the absence of this DC subset resulted in complete resistance to experimental CM. This was accompanied with dramatic reduction of brain CD8(+) T cells, and those few cerebral CD8(+) T cells present had a less activated phenotype, unlike their wildtype counterparts that expressed IFN-γ and especially granzyme B. This almost complete absence of local cellular responses was also associated with reduced parasite load in the brain.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Lectins, C-Type/physiology , Malaria, Cerebral/immunology , Malaria, Cerebral/pathology , Receptors, Immunologic/physiology , Animals , CD11c Antigen/biosynthesis , Cell Death/immunology , Clone Cells , Dendritic Cells/parasitology , Diphtheria Toxin/administration & dosage , Diphtheria Toxin/toxicity , Disease Models, Animal , Disease Resistance/genetics , Disease Resistance/immunology , Female , Humans , Lectins, C-Type/biosynthesis , Malaria, Cerebral/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Plasmodium berghei/immunology , Receptors, Immunologic/biosynthesis
8.
Blood ; 118(25): 6591-600, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22021367

ABSTRACT

In addition to adaptive T cells, the thymus supports the development of unconventional T cells such as natural killer T (NKT) and CD8αα intraepithelial lymphocytes (IELs), which have innate functional properties, particular antigenic specificities, and tissue localization. Both conventional and innate T cells are believed to develop from common precursors undergoing instructive, TCR-mediated lineage fate decisions, but innate T cells are proposed to undergo positive instead of negative selection in response to agonistic TCR signals. In the present study, we show that, in contrast to conventional αßT cells, innate αßT cells are not selected against functional TCRγ rearrangements and express TCRγ mRNA. Likewise, in contrast to the majority of γδT cells, thymic innate γδT cells are not efficiently selected against functional TCRß chains. In precursors of conventional T cells, autonomous TCR signals emanating from the pre-TCR or γδTCR in the absence of ligand mediate selection against the TCR of the opposite isotype and αß/γδ lineage commitment. Our data suggest that developing innate T cells ignore such signals and rely solely on agonistic TCR interactions. Consistently, most innate T cells reacted strongly against autologous thymocytes. These results suggest that innate and adaptive T-cell lineages do not develop from the same pool of precursors and potentially diverge before αß/γδ lineage commitment.


Subject(s)
Cell Lineage/genetics , Precursor Cells, T-Lymphoid/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Adaptive Immunity , Animals , Antibody Diversity/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/immunology , Female , Flow Cytometry , Gene Expression , Gene Rearrangement, T-Lymphocyte/genetics , Immunity, Innate , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Precursor Cells, T-Lymphoid/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymocytes/immunology , Thymocytes/metabolism
9.
J Immunol ; 181(3): 1710-6, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18641307

ABSTRACT

Although there has been great progress in the characterization of alphabeta T cell differentiation, selection, and function, gammadelta T cells have remained poorly understood. One of the main reasons for this is the lack of gammadelta T cell-specific surface markers other than the TCR chains themselves. In this study we describe two novel surface receptors, SCART1 and SCART2. SCARTs are related to CD5, CD6, and CD163 scavenger receptors but, unlike them, are found primarily on developing and mature gammadelta T cells. Characterization of SCART2 positive immature and peripheral gammadelta T cells suggests that they undergo lineage specification in the thymus and belong to a new IL-17-producing subset with distinct homing capabilities.


Subject(s)
Aging/immunology , Cells/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , Cell Lineage/immunology , Cell Movement/immunology , Cells, Cultured , Dermis/cytology , Dermis/immunology , Down-Regulation , Interleukin-17/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Molecular Sequence Data , Phylogeny , Receptors, Antigen, T-Cell, gamma-delta/classification , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Sequence Alignment , Signal Transduction/immunology , Thymus Gland/immunology
10.
Int Immunol ; 20(9): 1211-8, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18644831

ABSTRACT

Extensive amplification of hematopoietic stem cells (HSCs) and their multipotent primitive progenitors (MPPs) in culture would greatly benefit not only clinical transplantation but also provide a potential tool to manipulate all cellular lineages derived from these cells for gene therapy and experimental purposes. Here, we demonstrate that mouse bone marrow cultures containing cells engineered to over-express NUP98-HOXB4 fusion protein support self-renewal of physiologically normal HSC and MPP for several weeks leading practically to their unlimited expansion. This allows time consuming and cumulative in vitro experimental manipulations without sacrificing their ability to differentiate in vivo or in vitro to any hematopoietic lineage.


Subject(s)
Bone Marrow Cells/cytology , Cell Line, Transformed , Genetic Engineering/methods , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/metabolism , Nuclear Pore Complex Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Transcription Factors/metabolism , Animals , Bone Marrow Cells/metabolism , Cell Differentiation , Cells, Cultured , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/genetics , Immune System , Mice , Mice, Congenic , Mice, Inbred C57BL , Nuclear Pore Complex Proteins/genetics , Recombinant Fusion Proteins/genetics , Retroviridae/genetics , Transcription Factors/genetics , Transduction, Genetic
11.
Front Immunol ; 9: 3043, 2018.
Article in English | MEDLINE | ID: mdl-30622538

ABSTRACT

Type 1 conventional CD103+ dendritic cells (cDC1) contribute significantly to the cytotoxic T lymphocyte (CTL) response during influenza virus infection; however, the mechanisms by which cDC1s promote CTL recruitment and viral clearance are unclear. We demonstrate that cDC1 ablation leads to a deficient influenza-specific primary CD8+ T cell response alongside severe pulmonary inflammation, intensifying susceptibility to infection. The diminished pulmonary CTL population is not only a consequence of reduced priming in the lymph node (LN), but also of dysregulated CD8+ T cell egression from the LN and reduced CD8+ T cell viability in the lungs. cDC1s promote S1PR expression on CTLs, a key chemokine receptor facilitating CTL LN egress, and express high levels of the T cell survival cytokine, IL-15, to support CTL viability at the site of infection. Moreover, cDC1 ablation leads to severe impairment of CD8+ T cell memory recall and cross-reactive protection, suggesting that cDC1 are not only involved in primary T cell activation, but also in supporting the development of effective memory CD8+ T cell precursors. Our findings demonstrate a previously unappreciated and multifaceted role of CD103+ DCs in controlling pulmonary T cell-mediated immune responses.


Subject(s)
Cell Communication/immunology , Dendritic Cells/immunology , Influenza, Human/immunology , T-Lymphocytes, Cytotoxic/immunology , Adoptive Transfer , Animals , Antigens, CD/metabolism , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Movement/immunology , Cell Survival/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Female , Humans , Immunization, Passive/methods , Immunologic Memory , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/blood , Influenza, Human/pathology , Influenza, Human/virology , Integrin alpha Chains/metabolism , Lung/cytology , Lung/immunology , Lung/pathology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/transplantation
12.
Cell Rep ; 25(11): 3099-3109.e3, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30540942

ABSTRACT

Inflammation-induced disappearance of tissue-resident macrophages represents a key pathogen defense mechanism. Using a model of systemic blood-stage malaria, we studied the dynamics of tissue-resident macrophages in multiple organs to determine how they are depleted and refilled during the course of disease. We show that Plasmodium infection results in a transient loss of embryonically established resident macrophages prior to the parasitemia peak. Fate-mapping analysis reveals that inflammatory monocytes contribute to the repopulation of the emptied niches of splenic red pulp macrophages and hepatic Kupffer cells, while lung alveolar macrophages refill their niche predominantly through self-renewal. Interestingly, the local microenvironment of the spleen and liver can "imprint" the molecular characteristics of fetal-derived macrophages on newly differentiated bone marrow-derived immigrants with remarkably similar gene expression profiles and turnover kinetics. Thus, the mononuclear phagocytic system has developed distinct but effective tissue-specific strategies to replenish emptied niches to guarantee the functional integrity of the system.


Subject(s)
Life Cycle Stages , Macrophages/parasitology , Malaria/parasitology , Organ Specificity , Animals , Bone Marrow Cells/pathology , Fetus/pathology , Inflammation/pathology , Kinetics , Kupffer Cells/pathology , Liver/pathology , Macrophage Activation , Macrophages/metabolism , Macrophages, Alveolar/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/pathology , Myeloid Cells/metabolism , Plasmodium/growth & development , Spleen/pathology , Transcriptome/genetics
13.
Nat Commun ; 9(1): 582, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29422500

ABSTRACT

Circulating CCR2+ monocytes are crucial for maintaining the adult tissue-resident F4/80hiMHCIIhi macrophage pool in the intestinal lamina propria. Here we show that a subpopulation of CCR2-independent F4/80hiMHCIIlow macrophages, which are the most abundant F4/80hi cells in neonates, gradually decline in number in adulthood; these macrophages likely represent the fetal contribution to F4/80hi cells. In colon adenomas of ApcMin/+ mice, F4/80hiMHCIIlow macrophages are not only preserved, but become the dominant subpopulation among tumour-resident macrophages during tumour progression. Furthermore, these pro-tumoural F4/80hiMHCIIlow and F4/80hiMHCIIhi macrophages can self-renew in the tumour and maintain their numbers mostly independent from bone marrow contribution. Analyses of colon adenomas indicate that CSF1 may be a key facilitator of macrophage self-renewal. In summary, the tumour microenvironment creates an isolated niche for tissue-resident macrophages that favours macrophage survival and self-renewal.


Subject(s)
Adenoma/immunology , Cell Self Renewal , Colonic Neoplasms/immunology , Colonic Polyps/immunology , Macrophages/cytology , Stem Cell Niche , Tumor Microenvironment , Adenoma/genetics , Adenomatous Polyposis Coli Protein/genetics , Animals , Antigens, Differentiation , Cell Survival , Colonic Neoplasms/genetics , Colonic Polyps/genetics , Histocompatibility Antigens Class II , Macrophage Colony-Stimulating Factor , Mice , Mice, Knockout , Mice, Transgenic , Neoplasms, Experimental , Receptors, CCR2/genetics
14.
Cell Rep ; 21(5): 1203-1214, 2017 Oct 31.
Article in English | MEDLINE | ID: mdl-29091760

ABSTRACT

Dendritic cells (DCs) and macrophages (Mϕs) share close developmental pathways and functional features, leading to blurring of the boundaries between these two cell lineages. However, a deeper understanding of DC and Mϕ ontogeny and more refined phenotypic and functional characterizations have helped to delineate pre-DC-derived conventional DCs (cDCs), including cDC1s and cDC2s, from monocyte-derived Mϕs. Here, we further refine DC/Mϕ cell classification and report that classically defined cDC2s contain a discrete population of monocyte-derived migratory antigen-presenting cells with Mϕ phenotype but functional DC features, including cross-presentation.


Subject(s)
Dendritic Cells/metabolism , Macrophages/metabolism , Monocytes/cytology , Animals , CD11c Antigen/metabolism , Cells, Cultured , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Dendritic Cells/classification , Dendritic Cells/cytology , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Lung/cytology , Lung/immunology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lymph Nodes/cytology , Lymph Nodes/immunology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/classification , Macrophages/cytology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Receptors, IgG/metabolism , Sialic Acid Binding Ig-like Lectin 1/metabolism , Spleen/cytology , Spleen/immunology
15.
Structure ; 11(9): 1151-61, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12962633

ABSTRACT

Due to a paucity of studies that synthesize structural, energetic, and functional analyses of a series of protein complexes representing distinct stages in an affinity maturation pathway, the biophysical basis for the molecular evolution of protein-protein interactions is poorly understood. Here, we combine crystal structures and binding-free energies of a series of variant superantigen (SAG)-major histocompatibility complex (MHC) class II complexes exhibiting increasingly higher affinity to reveal that this affinity maturation pathway is controlled largely by two biophysical factors: shape complementarity and buried hydrophobic surface. These factors, however, do not contribute equivalently to the affinity maturation of the interface, as the former dominates the early steps of the maturation process while the latter is responsible for improved binding in later steps. Functional assays reveal how affinity maturation of the SAG-MHC interface corresponds to T cell activation by SAGs.


Subject(s)
Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/metabolism , Protein Binding , Superantigens/chemistry , Superantigens/metabolism , Binding Sites , Enterotoxins/chemistry , Enterotoxins/metabolism , HLA-DR1 Antigen/chemistry , HLA-DR1 Antigen/metabolism , Hydrophobic and Hydrophilic Interactions , Lymphocyte Activation , Models, Molecular , Protein Structure, Quaternary , Protein Structure, Tertiary , Signal Transduction , T-Lymphocytes/chemistry , T-Lymphocytes/metabolism
16.
Structure ; 10(5): 687-99, 2002 May.
Article in English | MEDLINE | ID: mdl-12015151

ABSTRACT

Superantigens (SAGs) crosslink MHC class II and TCR molecules, resulting in an overstimulation of T cells associated with human disease. SAGs interact with several different surfaces on MHC molecules, necessitating the formation of multiple distinct MHC-SAG-TCR ternary signaling complexes. Variability in SAG-TCR binding modes could also contribute to the structural heterogeneity of SAG-dependent signaling complexes. We report crystal structures of the streptococcal SAGs SpeA and SpeC in complex with their corresponding TCR beta chain ligands that reveal distinct TCR binding modes. The SpeC-TCR beta chain complex structure, coupled with the recently determined SpeC-HLA-DR2a complex structure, provides a model for a novel T cell signaling complex that precludes direct TCR-MHC interactions. Thus, highly efficient T cell activation may be achieved through structurally diverse strategies of TCR ligation.


Subject(s)
Bacterial Proteins/chemistry , Exotoxins/chemistry , Membrane Proteins/chemistry , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Signal Transduction/physiology , Streptococcus pyogenes/chemistry , Superantigens/chemistry , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Exotoxins/genetics , Exotoxins/metabolism , Genes, MHC Class II , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Superantigens/genetics , Superantigens/metabolism , T-Lymphocytes/chemistry , T-Lymphocytes/metabolism
17.
Cell Rep ; 16(6): 1749-1761, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27477286

ABSTRACT

Tissue macrophages exhibit diverse functions, ranging from the maintenance of tissue homeostasis, including clearance of senescent erythrocytes and cell debris, to modulation of inflammation and immunity. Their contribution to the control of blood-stage malaria remains unclear. Here, we show that in the absence of tissue-resident CD169(+) macrophages, Plasmodium berghei ANKA (PbA) infection results in significantly increased parasite sequestration, leading to vascular occlusion and leakage and augmented tissue deposition of the malarial pigment hemozoin. This leads to widespread tissue damage culminating in multiple organ inflammation. Thus, the capacity of CD169(+) macrophages to contain the parasite burden and its sequestration into different tissues and to limit infection-induced inflammation is crucial to mitigating Plasmodium infection and pathogenesis.


Subject(s)
Macrophages/parasitology , Malaria/immunology , Plasmodium berghei/parasitology , Sialic Acid Binding Ig-like Lectin 1/genetics , Animals , Erythrocytes/parasitology , Hemeproteins/metabolism , Macrophages/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic
18.
Immunol Lett ; 160(2): 113-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24472602

ABSTRACT

CD19 plays a crucial role in mature B cell development as best exemplified by the finding that CD19 deficient mice have severely reduced mature B cell compartments (Engel et al., 1995; Rickert et al., 1995). In the present study we show that the transition into the mature B cell compartments is heavily dependent on the correct amount of CD19 expression. Thus, Nup-98-HoxB4 immortalized hematopoietic stem cells (HSCs) over-expressing CD19 show upon transplantation an impaired pro/pre B to immature B cell transition in the bone marrow, whereas Nup-98-HoxB4 HSCs expressing a shRNA that down-modulates CD19 expression show upon transplantation a strongly reduced mature B cell compartment. Overall our findings indicate that too high CD19 expression might result into too strong BCR signaling in the bone marrow and therefore causing negative selection. Too low CD19 expression might result into too little BCR signaling and thereby preventing the B cells to enter the mature pool (absence of positive selection).


Subject(s)
Antigens, CD19/genetics , Bone Marrow Cells/metabolism , Hematopoietic Stem Cells/metabolism , Pre-B Cell Receptors/genetics , Precursor Cells, B-Lymphoid/metabolism , Animals , Antigens, CD19/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Differentiation , Female , Gene Expression Regulation , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Immunophenotyping , Male , Mice , Mice, Knockout , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/immunology , Pre-B Cell Receptors/immunology , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Signal Transduction , Transcription Factors/genetics , Transcription Factors/immunology
19.
Stem Cells Dev ; 20(7): 1131-42, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21348597

ABSTRACT

Induced pluripotent stem (iPS) cells have been generated from bone marrow (BM) hematopoietic progenitor cells by ectopic expression of Sox-2, Oct-4, and Klf-4 with the hope that they may differentiate more efficiently than embryonic stem (ES) cells in vitro into hematopoietic cell lineages because of their epigenetic memory. An in vitro culture system has been standardized to allow a quantitative assessment of the capacities of different ES, BM-derived iPS, and fibroblast-derived iPS cell lines developing to erythroid, myeloid, and lymphoid cell lineages. Surprisingly, the efficiency to differentiate BM-derived iPS cells to hematopoietic cells in vitro is severely reduced compared with ES cells and fibroblast-derived iPS cells. Undifferentiated as well as differentiated stages of the BM-derived iPS lines express elevated mRNA levels of the transcription factors Sox-2, Oct-4, and Klf-4 with which the iPS cells have been transduced. Overexpression of the transcription factors inhibits development of Flk-1(+) mesodermal to CD45(+) hematopoietic progenitors. The overexpression of Sox-2 appears to be inversely related to hematogenic potency. These results suggest that iPS cell generation with the aim of developing hematopoietic cells should be controlled and selected for low levels of transduced Sox-2, Oct-4, and Kfl-4 expression.


Subject(s)
Cell Differentiation , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Transcription Factors/metabolism , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Cell Line , Cell Proliferation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Flow Cytometry/methods , Gene Expression , Genetic Vectors , Germ Layers/cytology , Germ Layers/metabolism , Hematopoietic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Octamer Transcription Factor-3/genetics , SOXB1 Transcription Factors/genetics , Teratoma/metabolism , Teratoma/secondary
20.
J Immunol ; 179(2): 1022-9, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17617594

ABSTRACT

The killer cell lectin-like receptor G1 (KLRG1) is expressed by NK cells and memory T cells in man and mice. Cadherins were recently identified as ligands for mouse KLRG1 but ligands for human KLRG1 have not yet been defined. In this study, we first demonstrate that human E-cadherin is a ligand for human KLRG1. This finding is remarkable because human and mouse KLRG1 show only an intermediate degree of homology (57% aa identity). In addition, we show that E-cadherin, expressed on K562 target cells, inhibited polyclonal human NK cells. Inhibition of NK cell function was observed consistently in three independent functional assays but the extent of inhibition was modest and required high expression of E-cadherin on target cells. E-cadherin function is often inactivated during development of human carcinomas and splice-site mutations resulting in in-frame loss of exon 8 or 9 occur frequently in diffuse type gastric carcinomas. Our experiments further revealed that interaction of human KLRG1 to E-cadherin was susceptible to these tumor-associated mutations and that KLRG1(+) NK cells were triggered more easily by K562 target cells carrying these mutations in comparison to target cells expressing wild-type E-cadherin. These results also indicate that the E-cadherin binding sites important for homophilic interaction are also involved in KLRG1 binding. Taken together, these data demonstrate that the main adhesion molecule of epithelial tissue, E-cadherin, is involved in regulation of NK cells in both humans and mice.


Subject(s)
Cadherins/genetics , Cadherins/metabolism , Cell Communication/physiology , Killer Cells, Natural/metabolism , Lectins, C-Type/metabolism , Trans-Activators/metabolism , Animals , Carcinoma/genetics , Coculture Techniques , Humans , Killer Cells, Natural/immunology , Mice , Mutation , Receptors, Immunologic , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL