Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
3.
Eur J Immunol ; 43(8): 2010-22, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23630120

ABSTRACT

Traumatic brain injury (TBI) elicits innate inflammatory responses that can lead to secondary brain injury. To better understand the mechanisms involved in TBI-induced inflammation, we examined the nature of macrophages responding to TBI in mice. In this model, brain macrophages were increased >20-fold the day after injury and >77-fold 4 days after injury in the ipsilateral hemisphere compared with sham controls. TBI macrophage subsets were identified by using a reporter mouse strain (YARG) that expresses eYFP from an internal ribosome entry site (IRES) inserted at the 3' end of the gene for arginase-1 (Arg1), a hallmark of alternatively activated (M2) macrophages. One day after TBI, 21 ± 1.5% of ipsilateral brain macrophages expressed relatively high levels of Arg1 as detected by yellow fluorescent protein, and this subpopulation declined thereafter. Arg1(+) cells localized with macrophages near the TBI lesion. Gene expression analysis of sorted Arg1(+) and Arg1(-) brain macrophages revealed that both populations had profiles that included features of conventional M2 macrophages and classically activated (M1) macrophages. The Arg1(+) cells differed from Arg1(-) cells in multiple aspects, most notably in their chemokine repertoires. Thus, the macrophage response to TBI initially involves heterogeneous polarization toward at least two major subsets.


Subject(s)
Arginase/metabolism , Brain Injuries/immunology , Brain/immunology , Macrophage Activation/immunology , Macrophages/immunology , Animals , Arginase/genetics , Bacterial Proteins/genetics , Cell Movement , Chemokines/biosynthesis , Gene Expression Profiling , Inflammation/immunology , Luminescent Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ribosomes/genetics , Ribosomes/metabolism
4.
PLoS One ; 6(8): e23800, 2011.
Article in English | MEDLINE | ID: mdl-21886823

ABSTRACT

Ferritin binds specifically and saturably to a variety of cell types, and recently several ferritin receptors have been cloned. TIM-2 is a specific receptor for H ferritin (HFt) in the mouse. TIM-2 is a member of the T cell immunoglobulin and mucin domain containing (TIM) protein family and plays an important role in immunity. The expression of TIM-2 outside of the immune system indicates that this receptor may have broader roles. We tested whether ferritin binding to TIM-2 can serve as an iron delivery mechanism. TIM-2 was transfected into normal (TCMK-1) mouse kidney cells, where it was appropriately expressed on the cell surface. HFt was labeled with (55)Fe and (55)Fe-HFt was incubated with TIM-2 positive cells or controls. (55)Fe-HFt uptake was observed only in TIM-2 positive cells. HFt uptake was also seen in A20 B cells, which express endogenous TIM-2. TIM-2 levels were not increased by iron chelation. Uptake of (55)Fe-HFt was specific and temperature-dependent. HFt taken up by TIM-2 positive cells transited through the endosome and eventually entered a lysosomal compartment, distinguishing the HFt pathway from that of transferrin, the classical vehicle for cellular iron delivery. Iron delivered following binding of HFt to TIM-2 entered the cytosol and became metabolically available, resulting in increased levels of endogenous intracellular ferritin. We conclude that TIM-2 can function as an iron uptake pathway.


Subject(s)
Apoferritins/metabolism , Iron/metabolism , Membrane Proteins/metabolism , Animals , Biological Transport , Endosomes/metabolism , Kidney/metabolism , Lysosomes/metabolism , Membrane Proteins/administration & dosage , Membrane Proteins/genetics , Mice , Protein Binding , Transfection
5.
J Biol Chem ; 286(2): 942-51, 2011 Jan 14.
Article in English | MEDLINE | ID: mdl-21036901

ABSTRACT

Cross-talk between Gα(i)- and Gα(q)-linked G-protein-coupled receptors yields synergistic Ca(2+) responses in a variety of cell types. Prior studies have shown that synergistic Ca(2+) responses from macrophage G-protein-coupled receptors are primarily dependent on phospholipase Cß3 (PLCß3), with a possible contribution of PLCß2, whereas signaling through PLCß4 interferes with synergy. We here show that synergy can be induced by the combination of Gßγ and Gα(q) activation of a single PLCß isoform. Synergy was absent in macrophages lacking both PLCß2 and PLCß3, but it was fully reconstituted following transduction with PLCß3 alone. Mechanisms of PLCß-mediated synergy were further explored in NIH-3T3 cells, which express little if any PLCß2. RNAi-mediated knockdown of endogenous PLCßs demonstrated that synergy in these cells was dependent on PLCß3, but PLCß1 and PLCß4 did not contribute, and overexpression of either isoform inhibited Ca(2+) synergy. When synergy was blocked by RNAi of endogenous PLCß3, it could be reconstituted by expression of either human PLCß3 or mouse PLCß2. In contrast, it could not be reconstituted by human PLCß3 with a mutation of the Y box, which disrupted activation by Gßγ, and it was only partially restored by human PLCß3 with a mutation of the C terminus, which partly disrupted activation by Gα(q). Thus, both Gßγ and Gα(q) contribute to activation of PLCß3 in cells for Ca(2+) synergy. We conclude that Ca(2+) synergy between Gα(i)-coupled and Gα(q)-coupled receptors requires the direct action of both Gßγ and Gα(q) on PLCß and is mediated primarily by PLCß3, although PLCß2 is also competent.


Subject(s)
Calcium Signaling/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Phospholipase C beta/metabolism , Animals , Complement C5a/metabolism , Humans , Macrophages/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mutagenesis , NIH 3T3 Cells , Phospholipase C beta/genetics , RNA, Small Interfering , Receptors, Purinergic P2/metabolism , Uridine Diphosphate/metabolism
6.
J Leukoc Biol ; 87(6): 1041-57, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20200401

ABSTRACT

Clostridium difficile toxins cause acute colitis by disrupting the enterocyte barrier and promoting inflammation. ToxB from C. difficile inactivates Rho family GTPases and causes release of cytokines and eicosanoids by macrophages. We studied the effects of ToxB on GPCR signaling in murine RAW264.7 macrophages and found that ToxB elevated Ca(2+) responses to Galphai-linked receptors, including the C5aR, but reduced responses to Galphaq-linked receptors, including the UDP receptors. Other Rho inhibitors also reduced UDP Ca(2+) responses, but they did not affect C5a responses, suggesting that ToxB inhibited UDP responses by inhibiting Rho but enhanced C5a responses by other mechanisms. By using PLCbeta isoform-deficient BMDM, we found that ToxB inhibited Ca(2+) signaling through PLCbeta4 but enhanced signaling through PLCbeta3. Effects of ToxB on GPCR Ca(2+) responses correlated with GPCR use of PLCbeta3 versus PLCbeta4. ToxB inhibited UDP Ca(2+) signaling without reducing InsP3 production or the sensitivity of cellular Ca(2+) stores to exogenous InsP3, suggesting that ToxB impairs UDP signaling at the level of InsP3/Ca(2+)coupling. In contrast, ToxB elevated InsP3 production by C5a, and the enhancement of Ca(2+) signaling by C5a was prevented by inhibition of PLA(2) or 5-LOX but not COX, implicating LTs but not prostanoids in the mechanism. In sum, ToxB has opposing, independently regulated effects on Ca(2+) signaling by different GPCR-linked PLCbeta isoforms in macrophages.


Subject(s)
Bacterial Proteins/pharmacology , Bacterial Toxins/pharmacology , Calcium/metabolism , Macrophages/drug effects , Phospholipase C beta/physiology , Phospholipases A2/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Blotting, Western , Cells, Cultured , Complement C5a/pharmacology , Cytoskeleton/metabolism , Female , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositols/metabolism , Protein Isoforms , Signal Transduction , Uridine Diphosphate/pharmacology , rho GTP-Binding Proteins/genetics
7.
Proc Natl Acad Sci U S A ; 107(8): 3505-10, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20133674

ABSTRACT

Ferritin is a spherical molecule composed of 24 subunits of two types, ferritin H chain (FHC) and ferritin L chain (FLC). Ferritin stores iron within cells, but it also circulates and binds specifically and saturably to a variety of cell types. For most cell types, this binding can be mediated by ferritin composed only of FHC (HFt) but not by ferritin composed only of FLC (LFt), indicating that binding of ferritin to cells is mediated by FHC but not FLC. By using expression cloning, we identified human transferrin receptor-1 (TfR1) as an important receptor for HFt with little or no binding to LFt. In vitro, HFt can be precipitated by soluble TfR1, showing that this interaction is not dependent on other proteins. Binding of HFt to TfR1 is partially inhibited by diferric transferrin, but it is hindered little, if at all, by HFE. After binding of HFt to TfR1 on the cell surface, HFt enters both endosomes and lysosomes. TfR1 accounts for most, if not all, of the binding of HFt to mitogen-activated T and B cells, circulating reticulocytes, and all cell lines that we have studied. The demonstration that TfR1 can bind HFt as well as Tf raises the possibility that this dual receptor function may coordinate the processing and use of iron by these iron-binding molecules.


Subject(s)
Antigens, CD/metabolism , Apoferritins/metabolism , B-Lymphocytes/metabolism , Receptors, Transferrin/metabolism , T-Lymphocytes/metabolism , Antigens, CD/genetics , Cell Line , Cloning, Molecular , Endosomes/metabolism , Humans , Lysosomes/metabolism , Protein Binding , Receptors, Transferrin/genetics , Transferrin/metabolism
8.
J Neurochem ; 109(4): 1144-56, 2009 May.
Article in English | MEDLINE | ID: mdl-19302484

ABSTRACT

Following neuronal injury, microglia initiate repair by phagocytosing dead neurons without eliciting inflammation. Prior evidence indicates triggering receptor expressed by myeloid cells-2 (TREM2) promotes phagocytosis and retards inflammation. However, evidence that microglia and neurons directly interact through TREM2 to orchestrate microglial function is lacking. We here demonstrate that TREM2 interacts with endogenous ligands on neurons. Staining with TREM2-Fc identified TREM2 ligands (TREM2-L) on Neuro2A cells and on cultured cortical and dopamine neurons. Apoptosis greatly increased the expression of TREM2-L. Furthermore, apoptotic neurons stimulated TREM2 signaling, and an anti-TREM2 mAb blocked stimulation. To examine the interaction between TREM2 and TREM2-L in phagocytosis, we studied BV2 microglial cells and their engulfment of apoptotic Neuro2A. One of our anti-TREM2 mAb, but not others, reduced engulfment, suggesting the presence of a functional site on TREM2 interacting with neurons. Further, Chinese hamster ovary cells transfected with TREM2 conferred phagocytic activity of neuronal cells demonstrating that TREM2 is both required and sufficient for competent uptake of apoptotic neuronal cells. Finally, while TREM2-L are expressed on neurons, TREM2 is not; in the brain, it is found on microglia. TREM2 and TREM2-L form a receptor-ligand pair connecting microglia with apoptotic neurons, directing removal of damaged cells to allow repair.


Subject(s)
Apoptosis/physiology , Membrane Glycoproteins/physiology , Microglia/physiology , Neurons/physiology , Phagocytosis/physiology , Receptors, Immunologic/physiology , Animals , Antibodies/chemistry , CHO Cells , Cell Communication , Cell Separation , Cricetinae , Cricetulus , Lentivirus/genetics , Ligands , Male , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/physiology , RNA, Messenger/genetics , Receptors, Immunologic/immunology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Transfection
9.
J Cell Biol ; 184(2): 215-23, 2009 Jan 26.
Article in English | MEDLINE | ID: mdl-19171755

ABSTRACT

Phagocytosis, which is essential for the immune response to pathogens, is initiated by specific interactions between pathogens and cell surface receptors expressed by phagocytes. This study identifies triggering receptor expressed on myeloid cells 2 (TREM-2) and its signaling counterpart DAP12 as a molecular complex that promotes phagocytosis of bacteria. Expression of TREM-2-DAP12 enables nonphagocytic Chinese hamster ovary cells to internalize bacteria. This function depends on actin cytoskeleton dynamics and the activity of the small guanosine triphosphatases Rac and Cdc42. Internalization also requires src kinase activity and tyrosine phosphorylation. In bone marrow-derived macrophages, phagocytosis is decreased in the absence of DAP12 and can be restored by expression of TREM-2-DAP12. Depletion of TREM-2 inhibits both binding and uptake of bacteria. Finally, TREM-2-dependent phagocytosis is impaired in Syk-deficient macrophages. This study highlights a novel role for TREM-2-DAP12 in the immune response to bacterial pathogens.


Subject(s)
Bacterial Physiological Phenomena , Membrane Glycoproteins/metabolism , Phagocytosis/immunology , Receptors, Immunologic/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bacteria/immunology , Bacteria/metabolism , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Escherichia coli/metabolism , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Staphylococcus aureus/metabolism
10.
J Neurochem ; 107(6): 1495-505, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19014383

ABSTRACT

Oligodendrocytes stain more strongly for iron than any other cell in the CNS, and they require iron for the production of myelin. For most cell types transferrin is the major iron delivery protein, yet neither transferrin receptor protein nor mRNA are detectable in mature oligodendrocytes. Thus an alternative iron delivery mechanism must exist. Given the significant long term consequences of developmental iron deficiency and the iron requirements for normal myelination, identification of the iron delivery mechanism for oligodendrocytes is important. Previously we have reported that oligodendrocytes bind H-ferritin and that H-ferritin binds to white matter tracts in vivo. Recently, T cell immunoglobulin and mucin domain-containing protein-2 (Tim-2) was shown to bind and internalize H-ferritin. In the present study we show that Tim-2 is expressed on oligodendrocytes both in vivo and in vitro. Further, the onset of saturable H-ferritin binding in CG4 oligodendrocyte cell line is accompanied by Tim-2 expression. Application of a blocking antibody to the extracellular domain of Tim-2 significantly reduces H-ferritin binding to the differentiated CG4 cells and primary oligodendrocytes. Tim-2 expression on CG4 cells is responsive to iron; decreasing with iron loading and increasing with iron chelation. Taken together, these data provide compelling evidence that Tim-2 is the H-ferritin receptor on oligodendrocytes suggesting it is the primary mechanism for iron acquisition by these cells.


Subject(s)
Apoferritins/metabolism , Membrane Proteins/metabolism , Oligodendroglia/metabolism , Animals , Animals, Newborn , Antibodies/pharmacology , Binding, Competitive/drug effects , Binding, Competitive/physiology , Brain/cytology , Brain/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Deferoxamine/pharmacology , Gene Expression/drug effects , Gene Expression/physiology , Glial Fibrillary Acidic Protein/metabolism , Iron/pharmacology , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Protein Binding/drug effects , Protein Binding/physiology , Rats , Rats, Sprague-Dawley , Siderophores/pharmacology , Stem Cells/drug effects , Stem Cells/physiology
11.
Infect Immun ; 76(6): 2439-47, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18391000

ABSTRACT

Macrophage recognition of Salmonella enterica serovar Typhimurium leads to a cascade of signaling events, including the activation of Src family and Syk kinases and the production of reactive oxygen species (ROS), which are critical for host innate defense during early stages of bacterial infection. ROS production depends on the NADPH oxidase, but little is known about the innate immune receptors and proximal adapters that regulate Salmonella-induced ROS. Herein, we demonstrate that serovar Typhimurium induces ROS through a pathway that requires both triggering receptor expressed on myeloid cells 2 (TREM2) and DAP12. This pathway is highly analogous to the pathways utilized by Fc receptors and integrins to regulate ROS production. Oral infection of mice with serovar Typhimurium demonstrates that the DAP12-dependent pathway regulates cecal colonization during early stages of Salmonella infection. Thus, DAP12 is an important regulator of Salmonella-induced ROS production in macrophages, and TREM2 is essential for linking DAP12 to the innate response to serovar Typhimurium.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Immunity, Innate/physiology , Macrophages/immunology , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Salmonella typhimurium/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cecum/microbiology , Cecum/pathology , Cell Line , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/microbiology , Gene Expression Regulation , Macrophages/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Reactive Oxygen Species/metabolism , Receptors, Immunologic/genetics , Signal Transduction
12.
J Biol Chem ; 283(25): 17351-61, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18411281

ABSTRACT

Studies in fibroblasts, neurons, and platelets have demonstrated the integration of signals from different G protein-coupled receptors (GPCRs) in raising intracellular free Ca(2+). To study signal integration in macrophages, we screened RAW264.7 cells and bone marrow-derived macrophages (BMDM) for their Ca(2+) response to GPCR ligands. We found a synergistic response to complement component 5a (C5a) in combination with uridine 5'-diphosphate (UDP), platelet activating factor (PAF), or lysophosphatidic acid (LPA). The C5a response was Galpha(i)-dependent, whereas the UDP, PAF, and LPA responses were Galpha(q)-dependent. Synergy between C5a and UDP, mediated by the C5a and P2Y6 receptors, required dual receptor occupancy, and affected the initial release of Ca(2+) from intracellular stores as well as sustained Ca(2+) levels. C5a and UDP synergized in generating inositol 1,4,5-trisphosphate, suggesting synergy in activating phospholipase C (PLC) beta. Macrophages expressed transcripts for three PLCbeta isoforms (PLCbeta2, PLCbeta3, and PLCbeta4), but GPCR ligands selectively used these isoforms in Ca(2+) signaling. C5a predominantly used PLCbeta3, whereas UDP used PLCbeta3 but also PLCbeta4. Neither ligand required PLCbeta2. Synergy between C5a and UDP likewise depended primarily on PLCbeta3. Importantly, the Ca(2+) signaling deficiency observed in PLCbeta3-deficient BMDM was reversed by re-constitution with PLCbeta3. Neither phosphatidylinositol (PI) 3-kinase nor protein kinase C was required for synergy. In contrast to Ca(2+), PI 3-kinase activation by C5a was inhibited by UDP, as was macropinocytosis, which depends on PI 3-kinase. PLCbeta3 may thus provide a selective target for inhibiting Ca(2+) responses to mediators of inflammation, including C5a, UDP, PAF, and LPA.


Subject(s)
Calcium/metabolism , Complement C5a/chemistry , Macrophages/metabolism , Phospholipase C beta/metabolism , Uridine Diphosphate/chemistry , Animals , Humans , Kinetics , Ligands , Mice , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Pinocytosis , Protein Isoforms , Signal Transduction
13.
J Immunol ; 177(4): 2051-5, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16887962

ABSTRACT

DAP12 is an ITAM-containing adapter that associates with receptors in myeloid and NK cells. DAP12-associated receptors can give activation signals leading to cytokine production; however, in some situations, DAP12 inhibits cytokine production stimulated through TLRs and FcRs. Here we show that Triggering Receptor Expressed on Myeloid cells (TREM)-2 is responsible for the DAP12-mediated inhibition in mouse macrophages. A chimeric receptor composed of the extracellular domain of TREM-2 and the cytoplasmic domain of DAP12 inhibited the TLR- and FcR-induced TNF production of DAP12-deficient macrophages, whereas a TREM-1 chimera did not. In wild-type macrophages, TREM-2 knockdown increased TLR-induced TNF production. A TREM-2 Fc fusion protein bound to macrophages, indicating that macrophages express a TREM-2 ligand. Thus, the interaction of TREM-2 and its ligand results in an inhibitory signal that can reduce the inflammatory response.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Macrophages/immunology , Macrophages/metabolism , Membrane Glycoproteins/biosynthesis , Receptors, Fc/antagonists & inhibitors , Receptors, Immunologic/biosynthesis , Toll-Like Receptors/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Cells, Cultured , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Ligands , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Fc/biosynthesis , Receptors, Immunologic/physiology , Toll-Like Receptors/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis
14.
J Bone Miner Res ; 21(2): 237-45, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16418779

ABSTRACT

UNLABELLED: Deficiency of the signaling adapter protein DAP12 or its associated receptor TREM2 is associated with abnormal OC development in humans. Here we examine the role of TREM2 in mouse OC development and function, including migration and resorption in vitro. These results provide new evidence that TREM2 regulates OC function independent of its effects on multinucleated OC differentiation. INTRODUCTION: TREM2 (triggering receptor expressed in myeloid cells-2) associates with the signaling adapter DAP12 in osteoclasts (OCs). Genetic mutation or deletion of either the TYROBP (DAP12) or TREM2 gene is associated with the human disorder of brain and bone, Nasu-Hakola disease. We and others recently showed the critical requirement for immunoreceptor tyrosine-based activation motif (ITAM) signals through DAP12 and the Fc Receptor gamma chain (FcRgamma) during OC development. Here, we further define the role of TREM2 in OC differentiation and describe a role for TREM2 in OC migration and bone resorption. MATERIALS AND METHODS: We generated monoclonal anti-mouse TREM2 antibodies (mAb), analyzed pre-osteoclasts and mature OCs for TREM2 surface expression, and determined the effect of antibody ligation on in vitro OC differentiation, resorption, and migration. TREM2 RNA interference (RNAi) was used to disrupt expression of TREM2 in pre-osteoclasts. RESULTS: Using flow cytometry, our studies reveal that TREM2 is weakly expressed on C57BL/6 bone marrow macrophages (BMMs) and is upregulated during culture with RANKL and macrophage-colony stimulating factor (M-CSF). The expression of TREM2 is unaltered in DAP12-deficient OCs. Using C57BL/6 BMMs or RAW264.7 precursors, anti-TREM2 mAb treatment with RANKL and M-CSF enhances the formation of multinuclear TRACP+ OCs compared with control mAb treatment. In contrast, these agents have no effect on DAP12-deficient precursors. Monoclonal Ab blockade of TREM2 on OCs generated from C57BL/6 BMMs results in decreased resorption of artificial calcium-phosphate substrate and dentine. Reduction of TREM2 expression in RAW264.7 cells by RNAi results in loss of OC formation in response to RANKL and M-CSF. Anti-TREM2 cross-linking enhances migration of C57BL/6 OCs and RAW246.7 OCs in response to M-CSF. CONCLUSIONS: Our studies indicate that the TREM2 receptor regulates OC multinucleation as well as resorption and migration of mature OCs. Thus, TREM2-DAP12 signals regulate both OC formation and function.


Subject(s)
Bone Resorption , Chemotaxis , Membrane Glycoproteins/physiology , Osteoclasts/cytology , Osteoclasts/physiology , Receptors, Immunologic/physiology , Adaptor Proteins, Signal Transducing/physiology , Animals , Antibodies, Monoclonal/pharmacology , Bone Resorption/genetics , Carrier Proteins/pharmacology , Cell Differentiation/genetics , Cells, Cultured , Chemotaxis/genetics , Macrophage Colony-Stimulating Factor/pharmacology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Membrane Glycoproteins/pharmacology , Mice , Mice, Inbred C57BL , Osteoclasts/drug effects , RANK Ligand , RNA Interference , Receptor Activator of Nuclear Factor-kappa B , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/genetics
16.
J Exp Med ; 202(7): 955-65, 2005 Oct 03.
Article in English | MEDLINE | ID: mdl-16203866

ABSTRACT

T cell immunoglobulin-domain and mucin-domain (TIM) proteins constitute a receptor family that was identified first on kidney and liver cells; recently it was also shown to be expressed on T cells. TIM-1 and -3 receptors denote different subsets of T cells and have distinct regulatory effects on T cell function. Ferritin is a spherical protein complex that is formed by 24 subunits of H- and L-ferritin. Ferritin stores iron atoms intracellularly, but it also circulates. H-ferritin, but not L-ferritin, shows saturable binding to subsets of human T and B cells, and its expression is increased in response to inflammation. We demonstrate that mouse TIM-2 is expressed on all splenic B cells, with increased levels on germinal center B cells. TIM-2 also is expressed in the liver, especially in bile duct epithelial cells, and in renal tubule cells. We further demonstrate that TIM-2 is a receptor for H-ferritin, but not for L-ferritin, and expression of TIM-2 permits the cellular uptake of H-ferritin into endosomes. This is the first identification of a receptor for ferritin and reveals a new role for TIM-2.


Subject(s)
B-Lymphocytes/metabolism , Endocytosis/immunology , Ferritins/metabolism , Kidney/metabolism , Liver/metabolism , Membrane Proteins/metabolism , Animals , Antibodies, Monoclonal , Cell Line, Tumor , Cloning, Molecular , DNA Primers , Flow Cytometry , Fluorescent Antibody Technique , Green Fluorescent Proteins , Immunohistochemistry , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction
17.
Cancer Res ; 64(1): 117-27, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14729615

ABSTRACT

High-grade astrocytomas and glioblastomas are usually unresectable because they extensively invade surrounding brain tissue. Here, we report the expression and function of a receptor on many astrocytomas that may alter both the proliferative and invasive potential of these tumors. Signal regulatory protein (SIRP) alpha1 is an immunoglobulin superfamily transmembrane glycoprotein that is normally expressed in subsets of myeloid and neuronal cells. Transfection of many cell types with SIRPalpha1, including glioblastomas, has been shown to inhibit their proliferation in response to a range of growth factors. Furthermore, the expression of a murine SIRPalpha1 mutant has been shown to enhance cell adhesion and initial cell spreading but to inhibit cell extension and movement. The extracellular portion of SIRPalpha1 binds CD47 (integrin-associated protein), although this interaction is not required for integrin-mediated activation of SIRPalpha1. On phosphorylation, SIRPalpha1 recruits the tyrosine phosphatases SHP-1 and SHP-2, which are important in its functions. Although SHP-1 is uniquely expressed on hematopoietic cells, SHP-2 is ubiquitously expressed, so that SIRPalpha1 has the potential to function in many cell types, including astrocytomas. Because SIRPalpha1 regulates cell functions that may contribute to the malignancy of these tumors, we examined the expression of SIRPs in astrocytoma cell lines by flow cytometry using a monoclonal antibody against all SIRPs. Screening of nine cell lines revealed clear cell surface expression of SIRPs on five cell lines, whereas Northern blotting for SIRPalpha transcripts showed mRNA present in eight of nine cell lines. All nine cell lines expressed the ligand for SIRPalpha1, CD47. To further examine the expression and function of SIRPs, we studied the SF126 and U373MG astrocytoma cell lines, both of which express SIRPs, in greater detail. SIRP transcripts in these cells are identical in sequence to SIRPalpha1. The expressed deglycosylated protein is the same size as SIRPalpha1, but in the astrocytoma cells, it is underglycosylated compared with SIRPalpha1 produced in transfected Chinese hamster ovary cells. It is nonetheless still capable of binding soluble CD47. Moreover, SIRPalpha1 in each of the two cell lines recruited SHP-2 on phosphorylation, and SIRPalpha1 phosphorylation in cultured cells is CD47 dependent. Finally, examination of frozen sections from 10 primary brain tumor biopsies by immunohistochemistry revealed expression of SIRPs on seven of the specimens, some of which expressed high levels of SIRPs. Most of the tumors also expressed CD47. This is the first demonstration that astrocytomas can express SIRPalpha. Given the known role of SIRPalpha in regulating cell adhesion and responses to mitogenic growth factors, the expression of SIRPalpha1 on astrocytomas may be of considerable importance in brain tumor biology, and it offers the potential of a new avenue for therapeutic intervention.


Subject(s)
Antigens, Differentiation , Astrocytoma/genetics , Gene Expression Regulation, Neoplastic/genetics , Membrane Glycoproteins/genetics , Neural Cell Adhesion Molecule L1/genetics , Receptors, Immunologic/genetics , Base Sequence , Brain Neoplasms/genetics , DNA Primers , Flow Cytometry , Glycosylation , Humans , Immunohistochemistry , Membrane Glycoproteins/metabolism , Molecular Sequence Data , Neural Cell Adhesion Molecule L1/metabolism , Receptors, Immunologic/metabolism , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Vanadates/pharmacology
18.
J Immunol ; 171(12): 6541-8, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14662855

ABSTRACT

By homology to triggering receptor expressed by myeloid cells-2, we screened the mouse expressed sequence tag database and isolated a new single Ig domain receptor, which we have expressed and characterized. The receptor is most similar in sequence to the human CMRF-35 receptor, and thus we have named it CMRF-35-like molecule (CLM)-1. By screening the mouse genome, we determined that CLM-1 was part of a multigene family located on a small segment of mouse chromosome 11. Each contains a single Ig domain, and they are expressed mainly in cells of the myeloid lineage. CLM-1 contains multiple cytoplasmic tyrosine residues, including two that lie in consensus immunoreceptor tyrosine-based inhibitory motifs, and we demonstrate that CLM-1 can associate with Src-homology 2 containing phosphatase-1. Expression of CLM-1 mRNA is down-regulated by treatment with receptor activator of NF-kappaB ligand (RANKL), a cytokine that drives osteoclast formation. Furthermore, expression of CLM-1 in the osteoclastogenic cell line RAW (RAW.CLM-1) prevents osteoclastogenesis induced by RANKL and TGF-beta. RAW.CLM-1 cells fail to multinucleate and do not up-regulate calcitonin receptor, but they express tartrate-resistant acid phosphatase, cathepsin K, and beta(3) integrin, suggesting that osteoclastogenesis is blocked at a late-intermediate stage. Thus, we define a new family of myeloid receptors, and demonstrate that the first member of this family, CLM-1, is an inhibitory receptor, able to block osteoclastogenesis.


Subject(s)
Antigens, Surface/chemistry , Growth Inhibitors/physiology , Membrane Glycoproteins/chemistry , Osteoclasts/cytology , Osteoclasts/immunology , Receptors, Immunologic/chemistry , Receptors, Immunologic/physiology , Sequence Homology, Amino Acid , Amino Acid Sequence , Animals , Cell Differentiation/immunology , Cell Line , Cell Line, Tumor , Cloning, Molecular , Growth Inhibitors/chemistry , Growth Inhibitors/genetics , Immunoglobulins/chemistry , Intracellular Signaling Peptides and Proteins , Leukemia P388 , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Multigene Family/immunology , Myeloid Cells/metabolism , Protein Phosphatase 1 , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/metabolism , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/genetics
19.
J Immunol ; 171(2): 594-9, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12847223

ABSTRACT

We recently described the cloning of murine triggering receptor expressed by myeloid cells (TREM) 2, a single Ig domain DNAX adaptor protein 12-associated receptor expressed by cells of the myeloid lineage. In this study, we describe the identification of ligands for TREM-2 on both bacteria and mammalian cells. First, by using a TREM-2A/IgG1-Fc fusion protein, we demonstrate specific binding to a number of Gram-negative and Gram-positive bacteria and to yeast. Furthermore, we show that fluorescently labeled Escherichia coli and Staphylococcus aureus bind specifically to TREM-2-transfected cells. The binding of TREM-2A/Ig fusion protein to E. coli can be inhibited by the bacterial products LPS, lipoteichoic acid, and peptidoglycan. Additionally, binding can be inhibited by a number of other anionic carbohydrate molecules, including dextran sulfate, suggesting that ligand recognition is based partly on charge. Using a sensitive reporter assay, we demonstrate activation of a TREM-2A/CD3zeta chimeric receptor by both bacteria and dextran sulfate. Finally, we demonstrate binding of TREM-2A/Ig fusion to a series of human astrocytoma lines but not to a variety of other cell lines. The binding to astrocytomas, like binding to bacteria, is inhibited by anionic bacterial products, suggesting either a similar charge-based ligand recognition method or overlapping binding sites for recognition of self- and pathogen-expressed ligands.


Subject(s)
Receptors, Immunologic/metabolism , Animals , Anions , Astrocytoma/metabolism , Astrocytoma/microbiology , Bacterial Adhesion/drug effects , Bacterial Adhesion/genetics , Bacterial Adhesion/immunology , Binding, Competitive/genetics , Binding, Competitive/immunology , Dextran Sulfate/pharmacology , Gram-Negative Bacteria/physiology , Gram-Positive Bacteria/physiology , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Jurkat Cells , Leukemia P388 , Ligands , Lipopolysaccharides/pharmacology , Mice , Peptidoglycan/pharmacology , Protein Binding/drug effects , Protein Binding/genetics , Protein Binding/immunology , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/genetics , Receptors, Immunologic/physiology , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , Solubility , Teichoic Acids/pharmacology , Transfection , Tumor Cells, Cultured
20.
Nature ; 420(6916): 703-6, 2002 Dec 12.
Article in English | MEDLINE | ID: mdl-12478301

ABSTRACT

The Alliance for Cellular Signaling is a large-scale collaboration designed to answer global questions about signalling networks. Pathways will be studied intensively in two cells--B lymphocytes (the cells of the immune system) and cardiac myocytes--to facilitate quantitative modelling. One goal is to catalyse complementary research in individual laboratories; to facilitate this, all alliance data are freely available for use by the entire research community.


Subject(s)
B-Lymphocytes/physiology , Myocytes, Cardiac/physiology , Research Design , Research Personnel/organization & administration , Research/organization & administration , Signal Transduction , B-Lymphocytes/cytology , Cooperative Behavior , Databases, Factual , International Cooperation , Internet , Ligands , Models, Biological , Myocytes, Cardiac/cytology , United States , Workforce
SELECTION OF CITATIONS
SEARCH DETAIL
...