Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
2.
J Exp Med ; 221(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38722309

ABSTRACT

SYNTAXIN-11 (STX11) is a SNARE protein that mediates the fusion of cytotoxic granules with the plasma membrane at the immunological synapses of CD8 T or NK cells. Autosomal recessive inheritance of deleterious STX11 variants impairs cytotoxic granule exocytosis, causing familial hemophagocytic lymphohistiocytosis type 4 (FHL-4). In several FHL-4 patients, we also observed hypogammaglobulinemia, elevated frequencies of naive B cells, and increased double-negative DN2:DN1 B cell ratios, indicating a hitherto unrecognized role of STX11 in humoral immunity. Detailed analysis of Stx11-deficient mice revealed impaired CD4 T cell help for B cells, associated with disrupted germinal center formation, reduced isotype class switching, and low antibody avidity. Mechanistically, Stx11-/- CD4 T cells exhibit impaired membrane fusion leading to reduced CD107a and CD40L surface mobilization and diminished IL-2 and IL-10 secretion. Our findings highlight a critical role of STX11 in SNARE-mediated membrane trafficking and vesicle exocytosis in CD4 T cells, important for successful CD4 T cell-B cell interactions. Deficiency in STX11 impairs CD4 T cell-dependent B cell differentiation and humoral responses.


Subject(s)
B-Lymphocytes , CD4-Positive T-Lymphocytes , Qa-SNARE Proteins , Animals , Qa-SNARE Proteins/metabolism , Qa-SNARE Proteins/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Mice , Humans , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Lymphohistiocytosis, Hemophagocytic/immunology , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/metabolism , Mice, Knockout , Mice, Inbred C57BL , Female , Male , Germinal Center/immunology , Germinal Center/metabolism , Immunity, Humoral , Exocytosis
3.
Mucosal Immunol ; 17(1): 67-80, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37918715

ABSTRACT

Dysregulated B cell responses have been described in inflammatory bowel disease (IBD) patients; however, the role of B cells in IBD pathology remained incompletely understood. We here provide evidence for the detrimental role of activated B cells during the onset of autoimmune intestinal inflammation. Using Wiskott-Aldrich Syndrome interacting protein deficient (Wipf1-/-) mice as a mouse model of chronic colitis, we identified clusters of differentiation (CD)86 expression on activated B cells as a crucial factor exacerbating pro-inflammatory cytokine production of intestinal CD4 T cells. Depleting B cells through anti-CD20 antibody treatment or blocking costimulatory signals mediated by CD86 through cytotoxic T lymphocyte antigen-4-immunoglobulin (CTLA-4-Ig) diminished intestinal inflammation in our mouse model of chronic IBD at the onset of disease. This was due to a reduction in aberrant humoral immune responses and reduced CD4 T cell pro-inflammatory cytokine production, especially interferon-g (IFN-g) and granulocyte-macrophage colony-stimulating factor (GM-CSF). Interestingly, in addition to B cells isolated from the inflamed colon of Wipf1-/- mice, we also found CD86 mRNA and protein expression upregulated on activated B cells isolated from inflamed tissue of human patients with IBD. B cell activation and CD86 expression were boosted by soluble CD40L in vitro, which we found in the serum of mice and human patients with IBD. In summary, our data provides detailed insight into the contribution of B cells to intestinal inflammation, with implications for the treatment of IBD.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Humans , CD4-Positive T-Lymphocytes , Inflammation/metabolism , Intestinal Mucosa , Intestines/pathology
4.
J Hepatol ; 79(2): 296-313, 2023 08.
Article in English | MEDLINE | ID: mdl-37224925

ABSTRACT

BACKGROUND & AIMS: The progression of non-alcoholic steatohepatitis (NASH) to fibrosis and hepatocellular carcinoma (HCC) is aggravated by auto-aggressive T cells. The gut-liver axis contributes to NASH, but the mechanisms involved and the consequences for NASH-induced fibrosis and liver cancer remain unknown. We investigated the role of gastrointestinal B cells in the development of NASH, fibrosis and NASH-induced HCC. METHODS: C57BL/6J wild-type (WT), B cell-deficient and different immunoglobulin-deficient or transgenic mice were fed distinct NASH-inducing diets or standard chow for 6 or 12 months, whereafter NASH, fibrosis, and NASH-induced HCC were assessed and analysed. Specific pathogen-free/germ-free WT and µMT mice (containing B cells only in the gastrointestinal tract) were fed a choline-deficient high-fat diet, and treated with an anti-CD20 antibody, whereafter NASH and fibrosis were assessed. Tissue biopsy samples from patients with simple steatosis, NASH and cirrhosis were analysed to correlate the secretion of immunoglobulins to clinicopathological features. Flow cytometry, immunohistochemistry and single-cell RNA-sequencing analysis were performed in liver and gastrointestinal tissue to characterise immune cells in mice and humans. RESULTS: Activated intestinal B cells were increased in mouse and human NASH samples and licensed metabolic T-cell activation to induce NASH independently of antigen specificity and gut microbiota. Genetic or therapeutic depletion of systemic or gastrointestinal B cells prevented or reverted NASH and liver fibrosis. IgA secretion was necessary for fibrosis induction by activating CD11b+CCR2+F4/80+CD11c-FCGR1+ hepatic myeloid cells through an IgA-FcR signalling axis. Similarly, patients with NASH had increased numbers of activated intestinal B cells; additionally, we observed a positive correlation between IgA levels and activated FcRg+ hepatic myeloid cells, as well the extent of liver fibrosis. CONCLUSIONS: Intestinal B cells and the IgA-FcR signalling axis represent potential therapeutic targets for the treatment of NASH. IMPACT AND IMPLICATIONS: There is currently no effective treatment for non-alcoholic steatohepatitis (NASH), which is associated with a substantial healthcare burden and is a growing risk factor for hepatocellular carcinoma (HCC). We have previously shown that NASH is an auto-aggressive condition aggravated, amongst others, by T cells. Therefore, we hypothesized that B cells might have a role in disease induction and progression. Our present work highlights that B cells have a dual role in NASH pathogenesis, being implicated in the activation of auto-aggressive T cells and the development of fibrosis via activation of monocyte-derived macrophages by secreted immunoglobulins (e.g., IgA). Furthermore, we show that the absence of B cells prevented HCC development. B cell-intrinsic signalling pathways, secreted immunoglobulins, and interactions of B cells with other immune cells are potential targets for combinatorial NASH therapies against inflammation and fibrosis.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Microbiota , Non-alcoholic Fatty Liver Disease , Humans , Mice , Animals , Non-alcoholic Fatty Liver Disease/complications , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Mice, Inbred C57BL , Liver/pathology , Fibrosis , Liver Cirrhosis/complications , Mice, Transgenic , Immunoglobulin A/metabolism , Immunoglobulin A/pharmacology , Disease Models, Animal , Diet, High-Fat/adverse effects
5.
Npj Imaging ; 1(1): 3, 2023.
Article in English | MEDLINE | ID: mdl-38665236

ABSTRACT

Conventional histology, as well as immunohistochemistry or immunofluorescence, enables the study of morphological and phenotypical changes during tissue inflammation with single-cell accuracy. However, although highly specific, such techniques require multiple time-consuming steps to apply exogenous labels, which might result in morphological deviations from native tissue structures. Unlike these techniques, mid-infrared (mid-IR) microspectroscopy is a label-free optical imaging method that retrieves endogenous biomolecular contrast without altering the native composition of the samples. Nevertheless, due to the strong optical absorption of water in biological tissues, conventional mid-IR microspectroscopy has been limited to dried thin (5-10 µm) tissue preparations and, thus, it also requires time-consuming steps-comparable to conventional imaging techniques. Here, as a step towards label-free analytical histology of unprocessed tissues, we applied mid-IR optoacoustic microscopy (MiROM) to retrieve intrinsic molecular contrast by vibrational excitation and, simultaneously, to overcome water-tissue opacity of conventional mid-IR imaging in thick (mm range) tissues. In this proof-of-concept study, we demonstrated application of MiROM for the fast, label-free, non-destructive assessment of the hallmarks of inflammation in excised white adipose tissue; i.e., formation of crown-like structures and changes in adipocyte morphology.

6.
Int J Radiat Oncol Biol Phys ; 112(3): 747-758, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34619330

ABSTRACT

PURPOSE: Radiation-induced cognitive deficits have a severe negative impact on pediatric brain tumor patients. The severity of cognitive symptoms is related to the age of the child when radiation was applied, with the most severe effects seen in the youngest. Previous studies using whole-brain irradiation in mice confirmed these findings. To understand ipsilateral and contralateral changes in the hippocampus after partial-brain radiation therapy (PBRT) of the left hemisphere, we assessed the neuroplasticity and changes in the microvasculature of the irradiated and nonirradiated hippocampus in juvenile mice. METHODS AND MATERIALS: The left hemispheres of 5-week-old mice were irradiated with 2, 8, and 20 Gy and a fractionated dose of 8 Gy in 2 fractions using a computed tomography image guided small animal radiation research platform. Long-term potentiation (LTP) has been monitored ex vivo in the hippocampal cornu ammonis 1 (CA1) region and was assessed 3 days and 5 and 10 weeks after PBRT in both hemispheres and compared to a sham group. Irradiation effects on the hippocampus microvasculature were quantified by efficient tissue clearing and multiorgan volumetric imaging. RESULTS: LTP in irradiated hippocampal slices of juvenile mice declines 3 days after radiation, lasts up to 10 weeks in the irradiated part of the hippocampus, and correlates with a significantly reduced microvasculature length. Specifically, LTP inhibition is sustained in the irradiated (20 Gy, 8 Gy in 2 fractions, 8 Gy, 2 Gy) hippocampus, whereas the contralateral hippocampus remains unaffected after PBRT. LTP inhibition in the irradiated hemisphere after PBRT might be associated with an impaired microvascular network. CONCLUSION: PBRT induces a long-lasting impairment in neuroplasticity and the microvessel network of the irradiated hippocampus, whereas the contralateral hippocampus remains unaffected. These findings provide insight into the design of PBRT strategies to better protect the young developing brain from cognitive decline.


Subject(s)
Cognitive Dysfunction , Hippocampus , Animals , Brain , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/etiology , Hippocampus/diagnostic imaging , Hippocampus/pathology , Humans , Mice , Mice, Inbred C57BL , Neuronal Plasticity/radiation effects
7.
J Cell Sci ; 134(15)2021 08 01.
Article in English | MEDLINE | ID: mdl-34342351

ABSTRACT

A better understanding of cell-cell and cell-niche interactions is crucial to comprehend the complexity of inflammatory or pathophysiological scenarios such as tissue damage during viral infections, the tumour microenvironment and neuroinflammation. Optical clearing and 3D volumetric imaging of large tissue pieces or whole organs is a rapidly developing methodology that holds great promise for the in-depth study of cells in their natural surroundings. These methods have mostly been applied to image structural components such as endothelial cells and neuronal architecture. Recent work now highlights the possibility of studying immune cells in detail within their respective immune niches. This Review summarizes recent developments in tissue clearing methods and 3D imaging, with a focus on the localization and quantification of immune cells. We first provide background to the optical challenges involved and their solutions before discussing published protocols for tissue clearing, the limitations of 3D imaging of immune cells and image analysis. Furthermore, we highlight possible applications for tissue clearing and propose future developments for the analysis of immune cells within homeostatic or inflammatory immune niches.


Subject(s)
Endothelial Cells , Imaging, Three-Dimensional , Optical Imaging , Tumor Microenvironment
8.
Front Immunol ; 12: 670290, 2021.
Article in English | MEDLINE | ID: mdl-33936114

ABSTRACT

Humoral immunity is mainly mediated by a B cell population highly specialized to synthesize and secrete large quantities of antibodies - the antibody-secreting cells (ASC). In the gastrointestinal environment, a mixture of foreign antigens from the diet, commensal microbiota as well as occasional harmful pathogens lead to a constant differentiation of B cells into ASC. Due to this permanent immune response, more than 80% of mammalian ASC reside in the gut, of which most express immunoglobulin A (IgA). IgA antibodies contribute to intestinal homeostasis and can mediate protective immunity. Recent evidence points at a role for gut-derived ASC in modulating immune responses also outside of mucosal tissues. We here summarize recent evidence for wandering ASC, their antibodies and their involvement in systemic immune responses.


Subject(s)
Immunity, Humoral/immunology , Immunity, Mucosal/immunology , Immunoglobulin A/immunology , Intestinal Mucosa/immunology , Plasma Cells/immunology , Animals , Chemotaxis, Leukocyte/immunology , Humans
9.
Front Immunol ; 11: 599495, 2020.
Article in English | MEDLINE | ID: mdl-33569052

ABSTRACT

Spatial information of cells in their tissue microenvironment is necessary to understand the complexity of pathophysiological processes. Volumetric imaging of cleared organs provides this information; however, current protocols are often elaborate, expensive, and organ specific. We developed a simplified, cost-effective, non-hazardous approach for efficient tissue clearing and multi-organ volumetric imaging (EMOVI). EMOVI enabled multiplexed antibody-based immunolabeling, provided adequate tissue transparency, maintained cellular morphology and preserved fluorochromes. Exemplarily, EMOVI allowed the detection and quantification of scarce cell populations during pneumonitis. EMOVI also permitted histo-cytometric analysis of MHC-II expressing cells, revealing distinct populations surrounding or infiltrating glomeruli of nephritic kidneys. Using EMOVI, we found widefield microscopy with real-time computational clearing as a valuable option for rapid image acquisition and detection of rare cellular events in cleared organs. EMOVI has the potential to make tissue clearing and volumetric imaging of immune cells applicable for a broad audience by facilitating flexibility in organ, fluorochrome and microscopy usage.


Subject(s)
Imaging, Three-Dimensional , Kidney Glomerulus , Animals , Inflammation/immunology , Inflammation/pathology , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Mice , Microscopy, Confocal , Nephritis/immunology , Nephritis/pathology
10.
Front Immunol ; 9: 699, 2018.
Article in English | MEDLINE | ID: mdl-29713321

ABSTRACT

Conventional dendritic cells (cDCs) are versatile activators of immune responses that develop as part of the myeloid lineage downstream of hematopoietic stem cells. We have recently shown that in mice precursors of cDCs, but not of other leukocytes, are marked by expression of DNGR-1/CLEC9A. To genetically deplete DNGR-1-expressing cDC precursors and their progeny, we crossed Clec9a-Cre mice to Rosa-lox-STOP-lox-diphtheria toxin (DTA) mice. These mice develop signs of age-dependent myeloproliferative disease, as has been observed in other DC-deficient mouse models. However, despite efficient depletion of cDC progenitors in these mice, cells with phenotypic characteristics of cDCs populate the spleen. These cells are functionally and transcriptionally similar to cDCs in wild type control mice but show somatic rearrangements of Ig-heavy chain genes, characteristic of lymphoid origin cells. Our studies reveal a previously unappreciated developmental heterogeneity of cDCs and suggest that the lymphoid lineage can generate cells with features of cDCs when myeloid cDC progenitors are impaired.


Subject(s)
Dendritic Cells/immunology , Lectins, C-Type/immunology , Receptors, Immunologic/immunology , Animals , Dendritic Cells/drug effects , Diphtheria Toxin/pharmacology , Lectins, C-Type/genetics , Mice , Receptors, Immunologic/genetics
11.
Proc Natl Acad Sci U S A ; 115(12): 3120-3125, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29507226

ABSTRACT

The transcription factor Foxp1 is critical for early B cell development. Despite frequent deregulation of Foxp1 in B cell lymphoma, the physiological functions of Foxp1 in mature B cells remain unknown. Here, we used conditional gene targeting in the B cell lineage and report that Foxp1 disruption in developing and mature B cells results in reduced numbers and frequencies of follicular and B-1 B cells and in impaired antibody production upon T cell-independent immunization in vivo. Moreover, Foxp1-deficient B cells are impaired in survival even though they exhibit an increased capacity to proliferate. Transcriptional analysis identified defective expression of the prosurvival Bcl-2 family gene Bcl2l1 encoding Bcl-xl in Foxp1-deficient B cells, and we identified Foxp1 binding in the regulatory region of Bcl2l1 Transgenic overexpression of Bcl2 rescued the survival defect in Foxp1-deficient mature B cells in vivo and restored peripheral B cell numbers. Thus, our results identify Foxp1 as a physiological regulator of mature B cell survival mediated in part via the control of Bcl-xl expression and imply that this pathway might contribute to the pathogenic function of aberrant Foxp1 expression in lymphoma.


Subject(s)
B-Lymphocytes/classification , B-Lymphocytes/physiology , Forkhead Transcription Factors/metabolism , Repressor Proteins/metabolism , Animals , Antibodies/metabolism , Antigens, CD19/metabolism , Forkhead Transcription Factors/genetics , Mice , Mice, Knockout , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Repressor Proteins/genetics , T-Lymphocytes/physiology , bcl-X Protein/genetics , bcl-X Protein/metabolism
12.
J Immunol ; 199(5): 1682-1695, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28747344

ABSTRACT

Rho family GTPases regulate diverse cellular events, such as cell motility, polarity, and vesicle traffic. Although a wealth of data exists on the canonical Rho GTPases RhoA, Rac1, and Cdc42, several other family members remain poorly studied. In B cells, we recently demonstrated a critical role for Cdc42 in plasma cell differentiation. In this study, we focus on a close homolog of Cdc42, TC10 (also known as RhoQ), and investigate its physiological role in B cells. By generating a TC10-deficient mouse model, we show that despite reduced total B cell numbers, B cell development in these mice occurs normally through distinct developmental stages. Upon immunization, IgM levels were reduced and, upon viral infection, germinal center responses were defective in TC10-deficient mice. BCR signaling was mildly affected, whereas cell migration remained normal in TC10-deficient B cells. Furthermore, by generating a TC10/Cdc42 double knockout mouse model, we found that TC10 can compensate for the lack of Cdc42 in TLR-induced cell activation and proliferation, so the two proteins play partly redundant roles. Taken together, by combining in vivo and in vitro analysis using TC10-deficient mice, we define the poorly studied Rho GTPase TC10 as an immunomodulatory molecule playing a role in physiological B cell responses.


Subject(s)
B-Lymphocytes/immunology , Orthomyxoviridae Infections/immunology , Vaccinia/immunology , cdc42 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Germinal Center/immunology , Immunomodulation , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , cdc42 GTP-Binding Protein/genetics , rho GTP-Binding Proteins/genetics
13.
J Exp Med ; 212(1): 53-72, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25547673

ABSTRACT

The small Rho GTPase Cdc42, known to interact with Wiskott-Aldrich syndrome (WAS) protein, is an important regulator of actin remodeling. Here, we show that genetic ablation of Cdc42 exclusively in the B cell lineage is sufficient to render mice unable to mount antibody responses. Indeed Cdc42-deficient mice are incapable of forming germinal centers or generating plasma B cells upon either viral infection or immunization. Such severe immune deficiency is caused by multiple and profound B cell abnormalities, including early blocks during B cell development; impaired antigen-driven BCR signaling and actin remodeling; defective antigen presentation and in vivo interaction with T cells; and a severe B cell-intrinsic block in plasma cell differentiation. Thus, our study presents a new perspective on Cdc42 as key regulator of B cell physiology.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Immunity, Humoral/immunology , Orthomyxoviridae Infections/immunology , cdc42 GTP-Binding Protein/immunology , Animals , Antibody Formation/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/ultrastructure , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cells, Cultured , Flow Cytometry , Gene Expression/immunology , Germinal Center/immunology , Germinal Center/metabolism , Immunity, Humoral/genetics , Influenza A virus/immunology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/virology , Reverse Transcriptase Polymerase Chain Reaction , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism
14.
Eur J Immunol ; 41(11): 3176-86, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21830209

ABSTRACT

The relevance of direct inflammatory signals (signal 3) for the activation of memory CD8(+) T cells during recall responses is so far unknown. We therefore investigated the direct impact of IL-12 and type I IFN on the formation, recall potential and protective capacity of memory T cells. Using CD8(+) T cells deficient for IL-12 or type I IFN receptors in an adoptive transfer system, we generated memory populations after infection with vaccinia virus, lymphocytic choriomeningitis virus or Listeria monocytogenes. The results demonstrate that in the absence of signal 3 cytokines during primary infection, functional memory T cells were formed. After retransfer into naïve mice, signal 3-deficient memory T cells were able to specifically lyse target cells in vivo under non-infectious conditions. However, after reinfection, secondary effector CD8(+) T cells lacking signal 3 were impaired in expansion and protective capacity dependent on the nature of the pathogen. We conclude that memory CD8(+) T cells depend on a signal 3 for expansion, independent of signals obtained during priming, thereby being influenced by the pathogen-induced inflammatory milieu during secondary infection. In summary, our results reveal an essential role for direct inflammatory cytokine signaling in secondary T-cell responses.


Subject(s)
Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Listeriosis/immunology , Lymphocyte Activation/immunology , Signal Transduction/immunology , Vaccinia/immunology , Animals , Cell Separation , Cytokines/biosynthesis , Cytokines/immunology , Flow Cytometry , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL
15.
Eur J Immunol ; 40(12): 3450-7, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21080375

ABSTRACT

Trogocytosis describes the transfer of surface determinants between immune cells and has been implicated in immune regulation. Most findings are based on in vitro studies since in vivo trogocytosis of immune cells is difficult to detect under physiological conditions. We used low frequencies of memory P14 T cells to demonstrate that T cells perform trogocytosis in vivo if in contact with APC pulsed with GP33-peptide or expressing the antigen endogenously. Furthermore, in vivo trogocytosis of T cells is demonstrated during infections with lymphocytic choriomeningitis virus and vaccinia virus. Trogocytosis-positive T cells revealed higher expression of activation marker and cytokines, showing a more activated phenotype compared to trogocytosis-negative T cells.


Subject(s)
Antigen-Presenting Cells/metabolism , Arenaviridae Infections/immunology , Lymphocytic choriomeningitis virus/immunology , Membrane Fusion , T-Lymphocytes/metabolism , Adoptive Transfer , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/pathology , Antigens, Surface/metabolism , Antigens, Viral/immunology , Cells, Cultured , Glycoproteins/immunology , Lymphocyte Activation , Lymphocytic choriomeningitis virus/pathogenicity , Membrane Fusion/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , T-Lymphocytes/pathology , T-Lymphocytes/virology , Viral Proteins/immunology
16.
Eur J Immunol ; 39(7): 1774-83, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19548244

ABSTRACT

To study the role of IL-12 as a third signal for T-cell activation and differentiation in vivo, direct IL-12 signaling to CD8(+) T cells was analyzed in bacterial and viral infections using the P14 T-cell adoptive transfer model with CD8(+) T cells that lack the IL-12 receptor. Results indicate that CD8(+) T cells deficient in IL-12 signaling were impaired in clonal expansion after Listeria monocytogenes infection but not after infection with lymphocytic choriomeningitis virus, vaccinia virus or vesicular stomatitis virus. Although limited in clonal expansion after Listeria infection, CD8(+) T cells deficient in IL-12 signaling exhibited normal degranulation activity, cytolytic functions, and secretion of IFN-gamma and TNF-alpha. However, CD8(+) T cells lacking IL-12 signaling failed to up-regulate KLRG1 and to down-regulate CD127 in the context of Listeria but not viral infections. Thus, direct IL-12 signaling to CD8(+) T cells determines the cell fate decision between short-lived effector cells and memory precursor effector cells, which is dependent on pathogen-induced local cytokine milieu.


Subject(s)
Cell Differentiation/immunology , Listeriosis/immunology , Lymphocytic Choriomeningitis/immunology , T-Lymphocytes/cytology , Adoptive Transfer/methods , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/virology , Cell Line , Cell Line, Tumor , Cell Proliferation , Flow Cytometry , Immunologic Memory/immunology , Interferon-gamma/immunology , Interleukin-7 Receptor alpha Subunit/immunology , Lectins, C-Type , Listeriosis/microbiology , Listeriosis/therapy , Lymphocytic Choriomeningitis/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Immunologic/immunology , Receptors, Interleukin-12/deficiency , Receptors, Interleukin-12/genetics , Receptors, Interleukin-12/immunology , Signal Transduction/immunology , T-Lymphocytes/microbiology , T-Lymphocytes/virology , Time Factors , Tumor Necrosis Factor-alpha/immunology
17.
Cell Microbiol ; 9(2): 502-13, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16978234

ABSTRACT

Bacillus anthracis is a sporulating Gram-positive bacterium that causes the disease anthrax. The highly stable spore is the infectious form of the bacterium that first interacts with the prospective host, and thus the interaction between the host and spore is vital to the development of disease. We focused our study on the response of murine splenocytes to the B. anthracis spore by using paraformaldehyde-inactivated spores (FIS), a treatment that prevents germination and production of products associated with vegetative bacilli. We found that murine splenocytes produce IL-12 and IFN-gamma in response to FIS. The IL-12 was secreted by CD11b cells, which functioned to induce the production of IFN-gamma by CD49b (DX5) NK cells. The production of these cytokines by splenocytes was not dependent on TLR2, TLR4, TLR9, Nod1, or Nod2; however, it was dependent on the signalling adapter protein MyD88. Unlike splenocytes, Nod1- and Nod2-transfected HEK cells were activated by FIS. Both IL-12 and IFN-gamma secretion were inhibited by treatment with B. anthracis lethal toxin. These observations suggest that the innate immune system recognizes spores with a MyD88-dependent receptor (or receptors) and responds by secreting inflammatory cytokines, which may ultimately aid in resisting infection.


Subject(s)
Bacillus anthracis/physiology , Cytokines/metabolism , Myeloid Differentiation Factor 88/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Spleen/cytology , Spores, Bacterial/immunology , Animals , Cell Line , Cytokines/genetics , Humans , Mice , Myeloid Differentiation Factor 88/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...