Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Exp Med ; 203(11): 2551-62, 2006 Oct 30.
Article in English | MEDLINE | ID: mdl-17060474

ABSTRACT

B cell life depends critically on the cytokine B cell-activating factor of the tumor necrosis factor family (BAFF). Lack of BAFF signaling leads to B cell death and immunodeficiency. Excessive BAFF signaling promotes lupus-like autoimmunity. Despite the great importance of BAFF to B cell biology, its signaling mechanism is not well characterized. We show that BAFF initiates signaling and transcriptional programs, which support B cell survival, metabolic fitness, and readiness for antigen-induced proliferation. We further identify a BAFF-specific protein kinase C beta-Akt signaling axis, which provides a connection between BAFF and generic growth factor-induced cellular responses.


Subject(s)
B-Cell Activating Factor/physiology , B-Lymphocytes/metabolism , Protein Kinase C/physiology , Proto-Oncogene Proteins c-akt/physiology , Animals , B-Lymphocytes/enzymology , Cell Proliferation , Cell Survival/immunology , Cells, Cultured , Mice , Protein Kinase C/deficiency , Protein Kinase C/genetics , Protein Kinase C beta , Signal Transduction/immunology
2.
J Exp Med ; 195(12): 1647-52, 2002 Jun 17.
Article in English | MEDLINE | ID: mdl-12070292

ABSTRACT

Activation of the nuclear factor (NF)-kappaB transcription complex by signals derived from the surface expressed B cell antigen receptor controls B cell development, survival, and antigenic responses. Activation of NF-kappaB is critically dependent on serine phosphorylation of the IkappaB protein by the multi-component IkappaB kinase (IKK) containing two catalytic subunits (IKKalpha and IKKbeta) and one regulatory subunit (IKKgamma). Using mice deficient for protein kinase C beta (PKCbeta) we show an essential role of PKCbeta in the phosphorylation of IKKalpha and the subsequent activation of NF-kappaB in B cells. Defective IKKalpha phosphorylation correlates with impaired B cell antigen receptor-mediated induction of the pro-survival protein Bcl-xL. Lack of IKKalpha phosphorylation and defective NF-kappaB induction in the absence of PKCbeta explains the similarity in immunodeficiencies caused by PKCbeta or IKKalpha ablation in B cells. Furthermore, the well established functional cooperation between the protein tyrosine kinase Bruton's tyrosine kinase (Btk), which regulates the activity of NF-kappaB and PKCbeta, suggests PKCbeta as a likely serine/threonine kinase component of the Btk-dependent NF-kappaB activating signal transduction chain downstream of the BCR.


Subject(s)
B-Lymphocytes/metabolism , Isoenzymes/metabolism , NF-kappa B/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , B-Lymphocytes/enzymology , I-kappa B Kinase , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Kinase C beta
3.
J Exp Med ; 197(11): 1573-83, 2003 Jun 02.
Article in English | MEDLINE | ID: mdl-12782719

ABSTRACT

Interleukin-10 (IL-10) is a potent deactivator of myeloid cells that limits the intensity and duration of immune and inflammatory responses. The activity of IL-10 can be suppressed during inflammation, infection, or after allogeneic tissue transplantation. We investigated whether inflammatory factors suppress IL-10 activity at the level of signal transduction. Out of many factors tested, only ligation of Fc receptors by immune complexes inhibited IL-10 activation of the Jak-Stat signaling pathway. IL-10 signaling was suppressed in rheumatoid arthritis joint macrophages that are exposed to immune complexes in vivo. Activation of macrophages with interferon-gamma was required for Fc receptor-mediated suppression of IL-10 signaling, which resulted in diminished activation of IL-10-inducible genes and reversal of IL-10-dependent suppression of cytokine production. The mechanism of inhibition involved decreased cell surface IL-10 receptor expression and Jak1 activation and was dependent on protein kinase C delta. These results establish that IL-10 signaling is regulated during inflammation and identify Fc receptors and interferon-gamma as important regulators of IL-10 activity. Generation of macrophages refractory to IL-10 can contribute to pathogenesis of inflammatory and infectious diseases characterized by production of interferon-gamma and immune complexes.


Subject(s)
Arthritis, Rheumatoid/immunology , Interleukin-10/metabolism , Receptors, IgG/metabolism , Antigen-Antibody Complex/metabolism , Base Sequence , DNA/genetics , Humans , In Vitro Techniques , Interferon-gamma/metabolism , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Protein Kinase C/metabolism , Signal Transduction
4.
Nature ; 431(7007): 456-61, 2004 Sep 23.
Article in English | MEDLINE | ID: mdl-15361883

ABSTRACT

Approximately 65% of B cells generated in human bone marrow are potentially harmful autoreactive B cells. Most of these cells are clonally deleted in the bone marrow, while those autoreactive B cells that escape to the periphery are anergized or perish before becoming mature B cells. Escape of self-reactive B cells from tolerance permits production of pathogenic auto-antibodies; recent studies suggest that extended B lymphocyte survival is a cause of autoimmune disease in mice and humans. Here we report a mechanism for the regulation of peripheral B-cell survival by serine/threonine protein kinase Cdelta (PKCdelta): spontaneous death of resting B cells is regulated by nuclear localization of PKCdelta that contributes to phosphorylation of histone H2B at serine 14 (S14-H2B). We show that treatment of B cells with the potent B-cell survival factor BAFF ('B-cell-activating factor belonging to the TNF family') prevents nuclear accumulation of PKCdelta. Our data suggest the existence of a previously unknown BAFF-induced and PKCdelta-mediated nuclear signalling pathway which regulates B-cell survival.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Nucleus/metabolism , Membrane Proteins/metabolism , Protein Kinase C/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Animals , B-Cell Activating Factor , B-Lymphocytes/enzymology , Cell Death , Cell Nucleus/enzymology , Cell Survival , Cells, Cultured , Fibroblasts , Mice , Phosphorylation , Protein Kinase C/deficiency , Protein Kinase C/genetics , Protein Kinase C-delta
5.
Curr Opin Immunol ; 16(2): 251-5, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15023421

ABSTRACT

The survival of mature resting B cells in the periphery depends on signaling from the B-cell receptor (BCR) and the B-cell activating factor of the TNF family receptor (BAFF-R). Engagement of both receptors promotes NF-kappa B activity, which contributes to B-cell survival through different pathways. BCR signaling leads to activation of the inhibitor of NF-kappa B kinase (IKK) complex via Carma1, Bcl10 and MALT1, whereas BAFF-R engagement promotes processing of NF-kappa B2 protein p100, which is dependent on NF-kappa B-inducing kinase (NIK) and IKK alpha. Proximal signaling intermediates are potentially common to both pathways. We suggest that BCR and BAFF-R survival signaling are mutually dependent. In addition, we propose that BAFF-R signaling enhances the expression of survival genes through direct chromatin modifications in NF-kappa B target gene promoters.


Subject(s)
B-Lymphocytes/metabolism , Signal Transduction , Animals , B-Cell Activation Factor Receptor , Cell Survival , Membrane Proteins/metabolism , Mice , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor/metabolism
6.
Ann N Y Acad Sci ; 987: 125-34, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12727631

ABSTRACT

Protein kinase C (PKC) is a family of serine/threonine kinases which mediate essential cellular signals required for activation, proliferation, differentiation, and survival. Several PKC members are expressed in B lineage cells and activated by stimulation of the B cell receptor (BCR), thus suggesting a contribution of PKCs to the B cell-mediated immune response. To understand the individual roles of PKCs for B cell immunity, mice deficient for PKCbetaI/II (PKCbeta) or PKCdelta were analyzed. PKCbeta and PKCdelta play essential but distinctive roles in B cell immunity. In addition to its role in B cell activation and humoral immunity, PKCbeta was recently shown to control NF-kappaB activation and survival of mature B cells. PKCdelta on the other hand specifically regulates the induction of tolerance in self-reactive B cells. Thus, individual PCKs regulate B cell immunity specifically.


Subject(s)
B-Lymphocytes/immunology , Immunity, Cellular , Protein Kinase C/metabolism , Animals , Cell Survival/physiology , Mice , NF-kappa B/metabolism , Protein Kinase C/physiology
7.
J Exp Med ; 209(4): 661-9, 2012 Apr 09.
Article in English | MEDLINE | ID: mdl-22412156

ABSTRACT

Effective antiviral immunity depends on the ability of infected cells or cells triggered with virus-derived nucleic acids to produce type I interferon (IFN), which activates transcription of numerous antiviral genes. However, disproportionately strong or chronic IFN expression is a common cause of inflammatory and autoimmune diseases. We describe an epigenetic mechanism that determines cell type-specific differences in IFN and IFN-stimulated gene (ISG) expression in response to exogenous signals. We identify di-methylation of histone H3 at lysine 9 (H3K9me2) as a suppressor of IFN and IFN-inducible antiviral gene expression. We show that levels of H3K9me2 at IFN and ISG correlate inversely with the scope and amplitude of IFN and ISG expression in fibroblasts and dendritic cells. Accordingly, genetic ablation or pharmacological inactivation of lysine methyltransferase G9a, which is essential for the generation of H3K9me2, resulted in phenotypic conversion of fibroblasts into highly potent IFN-producing cells and rendered these cells resistant to pathogenic RNA viruses. In summary, our studies implicate H3K9me2 and enzymes controlling its abundance as key regulators of innate antiviral immunity.


Subject(s)
Epigenesis, Genetic , Histones/metabolism , Interferons/biosynthesis , Virus Diseases/immunology , Animals , Histone-Lysine N-Methyltransferase/physiology , Immunity, Innate , Methylation , Mice , Mice, Inbred C57BL
8.
Genes Dev ; 21(16): 1999-2004, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17626790

ABSTRACT

Binding of microRNA (miRNA) to mRNA within the RNA-induced silencing complex (RISC) leads to either translational inhibition or to destruction of the target mRNA. Both of these functions are executed by Argonaute 2 (Ago2). Using hematopoiesis in mice as a model system to study the physiological function of Ago2 in vivo, we found that Ago2 controls early development of lymphoid and erythroid cells. We show that the unique and defining feature of Ago2, the Slicer endonuclease activity, is dispensable for hematopoiesis. Instead, we identified Ago2 as a key regulator of miRNA homeostasis. Deficiency in Ago2 impairs miRNA biogenesis from precursor-miRNAs followed by a reduction in miRNA expression levels. Collectively, our data identify Ago2 as a highly specialized member of the Argonaute family with an essential nonredundant Slicer-independent function within the mammalian miRNA pathway.


Subject(s)
Eukaryotic Initiation Factor-2/physiology , Hematopoiesis/physiology , MicroRNAs/metabolism , Animals , Argonaute Proteins , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Line , Erythroblasts/cytology , Erythroblasts/metabolism , Erythropoiesis/genetics , Erythropoiesis/physiology , Eukaryotic Initiation Factor-2/deficiency , Eukaryotic Initiation Factor-2/genetics , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , In Vitro Techniques , Lymphopoiesis/genetics , Lymphopoiesis/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics
9.
Mol Cell ; 27(4): 596-608, 2007 Aug 17.
Article in English | MEDLINE | ID: mdl-17707231

ABSTRACT

Epigenetic gene silencing in eukaryotes is regulated in part by lysine methylation of the core histone proteins. While histone lysine methylation is known to control gene expression through the recruitment of modification-specific effector proteins, it remains unknown whether nonhistone chromatin proteins are targets for similar modification-recognition systems. Here we show that the histone H3 methyltransferase G9a contains a conserved methylation motif with marked sequence similarity to H3 itself. As with methylation of H3 lysine 9, autocatalytic G9a methylation is necessary and sufficient to mediate in vivo interaction with the epigenetic regulator heterochromatin protein 1 (HP1), and this methyl-dependent interaction can be reversed by adjacent G9a phosphorylation. NMR analysis indicates that the HP1 chromodomain recognizes methyl-G9a through a binding mode similar to that used in recognition of methyl-H3K9, demonstrating that the chromodomain functions as a generalized methyl-lysine binding module. These data reveal histone-like modification cassettes - or "histone mimics" - as a distinct class of nonhistone methylation targets and directly extend the principles of the histone code to the regulation of nonhistone proteins.


Subject(s)
DNA Methylation , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Molecular Mimicry , Multiprotein Complexes/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/chemistry , Humans , Lysine/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Methyltransferases
10.
J Immunol ; 178(12): 7814-21, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17548619

ABSTRACT

Lytic granule exocytosis is the major pathway used by CD8+ CTL to kill virally infected and tumor cells. Despite the obvious importance of this pathway in adaptive T cell immunity, the molecular identity of enzymes involved in the regulation of this process is poorly characterized. One signal known to be critical for the regulation of granule exocytosis-mediated cytotoxicity in CD8+ T cells is Ag receptor-induced activation of protein kinase C (PKC). However, it is not known which step of the process is regulated by PKC. In addition, it has not been determined to date which of the PKC family members is required for the regulation of lytic granule exocytosis. By combination of pharmacological inhibitors and use of mice with targeted gene deletions, we show that PKCdelta is required for granule exocytosis-mediated lytic function in mouse CD8+ T cells. Our studies demonstrate that PKCdelta is required for lytic granule exocytosis, but is dispensable for activation, cytokine production, and expression of cytolytic molecules in response to TCR stimulation. Importantly, defective lytic function in PKCdelta-deficient cytotoxic lymphocytes is reversed by ectopic expression of PKCdelta. Finally, we show that PKCdelta is not involved in target cell-induced reorientation of the microtubule-organizing center, but is required for the subsequent exocytosis step, i.e., lytic granule polarization. Thus, our studies identify PKCdelta as a novel and selective regulator of Ag receptor-induced lytic granule polarization in mouse CD8+ T cells.


Subject(s)
Cytoplasmic Granules/ultrastructure , Exocytosis/immunology , Protein Kinase C-delta/physiology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , CD8 Antigens/analysis , Cytoplasmic Granules/immunology , Exocytosis/genetics , Granzymes/metabolism , Mice , Mice, Mutant Strains , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/genetics , Receptors, Antigen, T-Cell/agonists , Sequence Deletion , T-Lymphocytes, Cytotoxic/enzymology , T-Lymphocytes, Cytotoxic/ultrastructure
11.
J Immunol ; 176(8): 4785-92, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16585572

ABSTRACT

Balanced activity of pro- and anti-inflammatory cytokines during innate immune responses is required to allow effective host defense while avoiding tissue damage and autoimmunity. Induction of cytokine production after recognition of pathogen-associated molecular patterns (PAMPs) by innate immune cells has been well demonstrated, but modulation of cytokine function by PAMPs is not well understood. In this study we show that stimulation of macrophages with zymosan, which contains PAMPs derived from yeast, rapidly extinguished macrophage responses to IL-10, a suppressive cytokine that limits inflammatory tissue damage but also compromises host defense. The mechanism of inhibition involved protein kinase Cbeta and internalization of IL-10R, and was independent of TLR2 and phagocytosis. Inhibition of IL-10 signaling and function required direct contact with zymosan, and cells in an inflammatory environment that had not contacted zymosan remained responsive to the paracrine activity of zymosan-induced IL-10. These results reveal a mechanism that regulates IL-10 function such that antimicrobial functions of infected macrophages are not suppressed, but the activation of surrounding noninfected cells and subsequent tissue damage are limited. The fate of individual cells in an inflammatory microenvironment is thus specified by dynamic interactions among host cells, microbes, and cytokines that determine the balance between protection and pathology.


Subject(s)
Interleukin-10/metabolism , Zymosan/pharmacology , Animals , CD11b Antigen/metabolism , Cells, Cultured , Down-Regulation/drug effects , Humans , Immunity, Innate , In Vitro Techniques , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Knockout , Opsonin Proteins/metabolism , Phagocytosis , Protein Kinase C/metabolism , Receptors, Interleukin/metabolism , Receptors, Interleukin-10 , Signal Transduction/drug effects , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Tumor Necrosis Factor-alpha/metabolism
12.
Proc Natl Acad Sci U S A ; 102(29): 10267-72, 2005 Jul 19.
Article in English | MEDLINE | ID: mdl-16000408

ABSTRACT

Cytokine signaling by the Jak-STAT pathway is subject to complex negative regulation that limits the amplitude and duration of signal transduction. Inhibition of signaling also mediates negative crosstalk, whereby factors with opposing biological activities crossinhibit each other's function. Here, we investigated a rapidly inducible mechanism that inhibited Jak-STAT activation by IFN-alpha, a cytokine that is important for antiviral responses, growth control, and modulation of immune responses. IFN-alpha-induced signaling and gene activation were inhibited by ligation of Fc receptors and Toll-like receptors 7 and 8 in a PKCbeta-dependent manner. Neither PKCbeta nor PKCdelta influenced responses of cells treated with IFN-alpha alone. Inhibition of IFN-alpha signaling correlated with suppression of IFN-alpha-dependent antiviral responses. PKC-mediated inhibition did not require de novo gene expression but involved the recruitment of PKCbeta to the IFN-alpha receptor and interaction with protein tyrosine phosphatase SHP-2, resulting in augmented phosphatase activity. PKC-mediated inhibition of IFN-alpha signaling was abolished in SHP-2-deficient cells, demonstrating a pivotal role for SHP-2 in this inhibitory pathway. Together, our data describe a rapidly inducible, direct mechanism of inhibition of Jak-STAT signaling mediated by a PKCbeta-SHP-2 signaling pathway.


Subject(s)
Gene Expression Regulation/immunology , Interferon-alpha/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Kinase C/metabolism , Protein Tyrosine Phosphatases/metabolism , Signal Transduction/immunology , Animals , Electrophoretic Mobility Shift Assay , Fluorescent Antibody Technique , Immunoblotting , Immunoprecipitation , Membrane Glycoproteins/metabolism , Mice , Microscopy, Confocal , NIH 3T3 Cells , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Receptors, Cell Surface/metabolism , Receptors, Fc/metabolism , Toll-Like Receptors , Transcriptional Activation
13.
Nature ; 416(6883): 860-5, 2002 Apr 25.
Article in English | MEDLINE | ID: mdl-11976686

ABSTRACT

Interaction of a B cell expressing self-specific B-cell antigen receptor (BCR) with an auto-antigen results in either clonal deletion or functional inactivation. Both of these processes lead to B-cell tolerance and are essential for the prevention of auto-immune diseases. Whereas clonal deletion results in the death of developing autoreactive B cells, functional inactivation of self-reactive B lymphocytes leads to complex changes in the phenotype of peripheral B cells, described collectively as anergy. Here we demonstrate that deficiency in protein kinase Cdelta (PKC-delta) prevents B-cell tolerance, and allows maturation and terminal differentiation of self-reactive B cells in the presence of the tolerizing antigen. The importance of PKC-delta in B-cell tolerance is further underscored by the appearance of autoreactive anti-DNA and anti-nuclear antibodies in the serum of PKC-delta-deficient mice. As deficiency of PKC-delta does not affect BCR-mediated B-cell activation in vitro and in vivo, our data suggest a selective and essential role of PKC-delta in tolerogenic, but not immunogenic, B-cell responses.


Subject(s)
Autoantigens/immunology , B-Lymphocytes/immunology , Immune Tolerance/immunology , Isoenzymes/metabolism , Protein Kinase C/metabolism , Animals , Antibodies, Antinuclear/blood , Antibodies, Antinuclear/immunology , Antigens, CD/metabolism , B-Lymphocytes/cytology , B7-2 Antigen , Calcium/metabolism , Calcium Signaling , Cell Differentiation , Clonal Anergy/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Deletion , Isoenzymes/deficiency , Isoenzymes/genetics , Isoenzymes/immunology , JNK Mitogen-Activated Protein Kinases , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Activation , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/deficiency , Protein Kinase C/genetics , Protein Kinase C/immunology , Protein Kinase C-delta , Signal Transduction , Spleen/cytology , Spleen/immunology
14.
Immunity ; 19(4): 621-32, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14563325

ABSTRACT

Bam32 is an adaptor protein recruited to the plasma membrane upon B cell receptor (BCR) crosslinking in a phosphoinositol 3-kinase (PI3K)-dependent manner; however, its physiologic function is unclear. To determine its physiologic function, we produced Bam32-deficient mice. Bam32(-/-) B cells develop normally but have impaired T-independent antibody responses in vivo and diminished responses to BCR crosslinking in vitro. Biochemical analysis revealed that Bam32 acts in a novel pathway leading from the BCR to MAPK/ERK Kinases (MEK1/2), MAPK/ERK Kinase Kinase-1 (MEKK1), extracellular signal-regulated kinase (ERK), and c-jun NH2-terminal kinase (JNK), but not p38 mitogen-activated protein kinase (p38). This pathway appears to be initiated by hematopoietic progenitor kinase-1 (HPK1), which interacts directly with Bam32, and differs from all previously characterized BCR signaling pathways in that it is required for normal BCR-mediated proliferation but not for B cell survival.


Subject(s)
Adaptor Proteins, Signal Transducing , B-Lymphocytes/physiology , Carrier Proteins/metabolism , Cell Division/physiology , Lipoproteins , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Receptors, Antigen, B-Cell/metabolism , Animals , JNK Mitogen-Activated Protein Kinases , Membrane Proteins/deficiency , Mice , Mice, Knockout
15.
J Biol Chem ; 279(29): 30123-32, 2004 Jul 16.
Article in English | MEDLINE | ID: mdl-15138267

ABSTRACT

The cAMP-response element-binding protein (CREB) is activated by phosphorylation on Ser-133 and plays a key role in the proliferative and survival responses of mature B cells to B cell receptor (BCR) signaling. The signal link between the BCR and CREB activation depends on a phorbol ester (phorbol 12-myristate 13-acetate)-sensitive protein kinase C (PKC) activity and not protein kinase A or calmodulin kinase; however, the identity and role of the PKC(s) activity has not been elucidated. We found the novel PKCdelta (nPKCdelta) activator bistratene A is sufficient to induce CREB phosphorylation in murine splenic B cells. The pharmacological inhibitor Gö6976, which targets conventional PKCs and PKCmu, has no effect on CREB phosphorylation, whereas the nPKCdelta inhibitor rottlerin blocks CREB phosphorylation following BCR cross-linking. Bryostatin 1 selectively prevents nPKCdelta depletion by phorbol 12-myristate 13-acetate when coapplied, coincident with protection of BCR-induced CREB phosphorylation. Ectopic expression of a kinase-inactive nPKCdelta blocks BCR-induced CREB phosphorylation in A20 B cells. In addition, BCR-induced CREB phosphorylation is significantly diminished in nPKCdelta-deficient splenic B cells in comparison with wild type mice. Consistent with the essential role for Bruton's tyrosine kinase and phospholipase Cgamma2 in mediating PKC activation, Bruton's tyrosine kinase- and phospholipase Cgamma2-deficient B cells display defective CREB phosphorylation by the BCR. We also found that p90 RSK directly phosphorylates CREB on Ser-133 following BCR cross-linking and is positioned downstream of nPKCdelta. Taken together, these results suggest a model in which BCR engagement leads to the phosphorylation of CREB via a signaling pathway that requires nPKCdelta and p90 RSK in mature B cells.


Subject(s)
B-Lymphocytes/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Protein Kinase C/metabolism , Receptors, Antigen, B-Cell/metabolism , Acetamides/pharmacology , Acetophenones/pharmacology , Animals , Benzopyrans/pharmacology , Binding Sites , Blotting, Western , Bryostatins , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Carbazoles/pharmacology , Cell Division , Cross-Linking Reagents/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Lactones/pharmacology , Macrolides , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Mitogens , Phosphorylation , Promoter Regions, Genetic , Protein Isoforms , Protein Kinase C-delta , Protein Structure, Tertiary , Pyrans/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Serine/chemistry , Signal Transduction , Spiro Compounds/pharmacology , Tetradecanoylphorbol Acetate , Time Factors
16.
J Immunol ; 172(10): 6476-81, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15128840

ABSTRACT

Interleukin-10 is a predominantly anti-inflammatory cytokine that inhibits macrophage and dendritic cell function, but can acquire proinflammatory activity during immune responses. We investigated whether type I IFNs, which are elevated during infections and in autoimmune diseases, modulate the activity of IL-10. Priming of primary human macrophages with low concentrations of IFN-alpha diminished the ability of IL-10 to suppress TNF-alpha production. IFN-alpha conferred a proinflammatory gain of function on IL-10, leading to IL-10 activation of expression of IFN-gamma-inducible, STAT1-dependent genes such as IFN regulatory factor 1, IFN-gamma-inducible protein-10 (CXCL10), and monokine induced by IFN-gamma (CXCL9). IFN-alpha priming resulted in greatly enhanced STAT1 activation in response to IL-10, and STAT1 was required for IL-10 activation of IFN-gamma-inducible protein-10 and monokine induced by IFN-gamma expression in IFN-alpha-primed cells. In control, unprimed cells, IL-10 activation of STAT1 was suppressed by constitutive activity of protein kinase C and Src homology 2 domain-containing phosphatase 1. These results demonstrate that type I IFNs regulate the balance between IL-10 anti- and proinflammatory activity, and provide insight into molecular mechanisms that regulate IL-10 function. Gain of IL-10 proinflammatory functions may contribute to its pathogenic role in autoimmune diseases characterized by elevated type I IFN levels, such as systemic lupus erythematosus.


Subject(s)
Inflammation Mediators/physiology , Interferon-alpha/immunology , Interleukin-10/physiology , Lupus Erythematosus, Systemic/immunology , Animals , Cells, Cultured , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drug Synergism , Humans , Inflammation Mediators/antagonists & inhibitors , Interferon-alpha/pharmacology , Interleukin-10/antagonists & inhibitors , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Lupus Erythematosus, Systemic/genetics , Macrophage Activation/immunology , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Mice, Mutant Strains , Protein Kinase C/metabolism , Protein Kinase C/physiology , Protein Kinase C-delta , Protein Phosphatase 1 , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/physiology , STAT1 Transcription Factor , Trans-Activators/antagonists & inhibitors , Trans-Activators/deficiency , Trans-Activators/genetics , Trans-Activators/metabolism , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL