Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 175
Filter
1.
J Immunol ; 180(3): 1729-36, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18209070

ABSTRACT

We evaluated the capacity of NK cells to influence expansion of CD4(+)CD25(+)FoxP3(+) regulatory T cells (Tregs) in response to microbial Ags, using Mycobacterium tuberculosis as a model. We previously found that Tregs expand when CD4(+) cells and monocytes are exposed to M. tuberculosis. Addition of NK cells that were activated by monokines (IL-12, IL-15, and IL-18) or by exposure to M. tuberculosis-stimulated monocytes reduced Treg expansion in response to M. tuberculosis. NK cell inhibition of Treg expansion was not mediated through IFN-gamma. Activated NK cells lysed expanded, but not freshly isolated Tregs. Although monokines increased NK cell expression of the activating receptors NKp46, NKG2D, 2B4, CD16, and DNAM-1, only anti-NKG2D and anti-NKp46 inhibited NK cell lysis of expanded Tregs. Of five NKG2D ligands, only UL16-binding protein 1 (ULBP1) was up-regulated on M. tuberculosis-expanded Tregs, and anti-ULBP1 inhibited NK cell lysis of expanded Tregs. M. tuberculosis-stimulated monocytes activated NK cells to lyse expanded Tregs, and this was also inhibited by anti-NKG2D and anti-ULBP1, confirming the physiological relevance of this effect. Our study identifies a potential new role for NK cells in maintaining the delicate balance between the regulatory and effector arms of the immune response.


Subject(s)
Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Mycobacterium tuberculosis/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/microbiology , CD4 Antigens/analysis , Cells, Cultured , Forkhead Transcription Factors/analysis , GPI-Linked Proteins , Histocompatibility Antigens Class I/metabolism , Humans , Interferon-gamma/metabolism , Interleukin-2 Receptor alpha Subunit/analysis , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Killer Cells, Natural/drug effects , Lymphocyte Activation , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Monokines/pharmacology , NK Cell Lectin-Like Receptor Subfamily K , Receptors, Immunologic/agonists , Receptors, Natural Killer Cell
2.
J Exp Med ; 204(10): 2397-405, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17875674

ABSTRACT

Monokines (i.e., interleukin [IL]-12, -18, and -15) induce natural killer (NK) cells to produce interferon-gamma (IFN-gamma), which is a critical factor for immune surveillance of cancer and monocyte clearance of infection. We show that SET, which is a potent inhibitor of protein phosphatase type 2A (PP2A) activity, is highly expressed in human CD56bright NK cells, which produce more IFN-gamma than CD56dim NK cells. SET was up-regulated upon monokine stimulation of primary human NK cells. Furthermore, ectopic overexpression of SET significantly enhanced IFN-gamma gene expression in monokine-stimulated NK cells. In contrast, RNAi-mediated suppression of SET expression renders NK cells inefficient in producing high levels of IFN-gamma in response to monokine costimulation. Mechanistically, suppression of PP2A activity by SET is important for IFN-gamma gene expression in NK cells. In fact, treatment of primary human NK cells with the PP2A activator 1,9-dideoxy-forskolin, as well as administration of the drug to C57BL/6 mice, significantly reduced NK-dependent IFN-gamma production in response to monokine treatment. Further, SET knockdown or pharmacologic activation of PP2A diminished extracellular signal-regulated kinase 1/2, p65RelA, signal transducer and activator of transduction 4 (STAT4), and STAT5 activity in monokine-stimulated NK cells, potentially contributing to the reduction in IFN-gamma gene expression. Thus, SET expression is essential for suppressing PP2A phosphatase activity that would otherwise limit NK cell antitumoral and/or antiinflammatory functions by impairing NK cell production of IFN-gamma.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Interferon-gamma/biosynthesis , Killer Cells, Natural/metabolism , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins , Enzyme Activation , Gene Expression Regulation , Histone Chaperones , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Monokines/pharmacology , Signal Transduction , Transcription Factors/genetics
3.
FEBS Lett ; 581(20): 3833-8, 2007 Aug 07.
Article in English | MEDLINE | ID: mdl-17659284

ABSTRACT

Glycogen storage disease type Ia (GSD-Ia) patients deficient in glucose-6-phosphatase-alpha manifest a disturbed glucose homeostasis. We hypothesized that disturbed glucose homeostasis might affect myeloid functions. Here, we show that GSD-Ia mice exhibit normal neutrophil activities but have elevated myeloid progenitor cells in the bone marrow and spleen. Interestingly, GSD-Ia mice exhibit a persistent increase in peripheral blood neutrophil counts along with elevated serum levels of granulocyte colony stimulating factor and cytokine-induced neutrophil chemoattractant. Taken together, our results suggest that a loss of glucose homeostasis can compromise the immune system, resulting in neutrophilia. This may explain some of the unexpected clinical manifestations seen in GSD-Ia.


Subject(s)
Cytokines/blood , Glycogen Storage Disease Type I/etiology , Glycogen Storage Disease Type I/metabolism , Neutrophils/metabolism , Animals , Calcium/analysis , Calcium/metabolism , Chemokine CXCL2 , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/physiology , Glycogen Storage Disease Type I/pathology , Mice , Mice, Knockout , Monokines/pharmacology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Respiratory Burst/physiology
4.
Protein Expr Purif ; 55(1): 132-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17513126

ABSTRACT

MIG (monokine induced by IFN-gamma) is a CXC-chemokine (CXCL9). It plays important roles in regulation of immune activities, and knowledge of the protein in areas of allograft transplants, autoimmune diseases, and cancer therapy is evolving quickly. The non-tagged recombinant murine MIG (rMuMIG) is therefore required to facilitate the functional studies of this important chemokine. Here we present the use of a bacteria expression system to produce non-tagged rMuMIG. The coding sequence for MIG was cloned into the pET28a (+) vector that was transformed into Escherichia coli BL21 (DE3). Expression of rMuMIG was induced by IPTG. Bacteria inclusion bodies containing the protein were isolated and washed to remove contaminated bacteria proteins, and resolved in Urea buffer. Renaturation of the denatured protein was carried out in the defined protein refolding buffer, and the refolded protein was purified using S-Sepharose cation exchange chromatography. The final preparation of the rMuMIG was more than 99% pure as measured by capillary electrophoresis and SDS-PAGE analysis. The biological activity of rMuMIG was demonstrated in a murine spleen cell chemotaxis assay with ED50 30 ng/ml. Further experiments showed that rMuMIG could inhibit proliferation of mouse bone marrow cells in vivo.


Subject(s)
Chemokines, CXC/biosynthesis , Monokines/biosynthesis , Recombinant Proteins/biosynthesis , Animals , Base Sequence , Bone Marrow Cells/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Chemokine CXCL9 , Chemokines, CXC/genetics , Chemokines, CXC/pharmacology , Chemotaxis , Electrophoresis, Capillary , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Interferon-gamma/pharmacology , Interferon-gamma/physiology , Mice , Molecular Sequence Data , Monokines/genetics , Monokines/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology
5.
J Cell Physiol ; 211(3): 630-7, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17192849

ABSTRACT

Human leucine zipper protein (LZIP) associates with CC chemokine receptor 1 (CCR1) and this protein-protein interaction should play an important role in leukocyte cell mobility. LZIP is known to regulate leukotactin-1 (Lkn-1)-dependent cell migration without affecting the chemotactic activities of other CC chemokines that bind to CCR1. Since Lkn-1 is engaged in the transcriptional activation of nuclear factor kappaB (NF-kappaB) and subsequent activation of the chemoattractant ability of leukocytes, we investigated the regulatory role of LZIP in the NF-kappaB pathway that is induced by CCR1-dependent chemokines. LZIP increased NF-kappaB-dependent luciferase activity in response to Lkn-1 in HOS/CCR1 cells and THP-1 cells. However, the NF-kappaB-dependent luciferase activities induced by other CCR1-dependent chemokines were not affected by LZIP overexpression. LZIP also increased Lkn-1-induced chemotactic activity through activation of the NF-kappaB pathway, whereas LZIP did not affect either the transactivation of NF-kappaB or the chemotactic activities induced by other CCR1-dependent chemokines. Western blot analysis showed that LZIP increased the degradation of IkappaBalpha induced by Lkn-1 but not by other CCR1-dependent chemokines. Results from electrophoretic mobility shift assay (EMSA) showed that LZIP enhanced the Lkn-1-induced DNA-binding activity of NF-kappaB. These data indicate that LZIP functions as a positive regulator in the NF-kappaB activation pathway that is triggered by Lkn-1 without affecting the transcriptional activation of NF-kappaB induced by other CCR1-dependent chemokines.


Subject(s)
Chemotaxis/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , NF-kappa B/metabolism , Receptors, Chemokine/metabolism , Signal Transduction/physiology , Cell Line, Tumor , Chemokine CCL11 , Chemokine CCL4 , Chemokine CCL5/metabolism , Chemokine CCL5/pharmacology , Chemokines, CC/metabolism , Chemokines, CC/pharmacology , Chemotaxis/drug effects , Cyclic AMP Response Element-Binding Protein/genetics , Humans , I-kappa B Proteins/metabolism , Luciferases/genetics , Macrophage Inflammatory Proteins/metabolism , Macrophage Inflammatory Proteins/pharmacology , Monocytes/cytology , Monocytes/metabolism , Monokines/metabolism , Monokines/pharmacology , NF-KappaB Inhibitor alpha , Osteosarcoma , RNA, Small Interfering , Receptors, CCR1 , Signal Transduction/drug effects
6.
Blood ; 109(6): 2481-7, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17110450

ABSTRACT

Natural killer (NK) cells contribute to host immunity, including tumor surveillance, through the production of interferon gamma (IFN-gamma). Although there is some knowledge about molecular mechanisms that induce IFN-gamma in NK cells, considerably less is known about the mechanisms that reduce its expression. Here, we investigate the role of the Hlx transcription factor in IFN-gamma production by NK cells. Hlx expression is induced in monokine-activated NK cells, but with delayed kinetics compared to IFN-gamma. Ectopic Hlx expression decreases IFN-gamma synthesis in primary human NK cells and IFN-gamma promoter activity in an NK-like cell line. Hlx protein levels inversely correlate with those of STAT4, a requisite factor for optimal IFN-gamma transcription. Mechanistically, we provide evidence indicating that Hlx overexpression accelerates dephosphorylation and proteasome-dependent degradation of the active Y693-phosphorylated form of STAT4. Thus, Hlx expression in activated NK cells temporally controls and limits the monokine-induced production of IFN-gamma, in part through the targeted depletion of STAT4.


Subject(s)
Down-Regulation , Homeodomain Proteins/metabolism , Interferon-gamma/biosynthesis , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Monokines/pharmacology , Transcription Factors/metabolism , Animals , Cell Line , Genes, Homeobox/genetics , Homeodomain Proteins/genetics , Humans , Interferon-gamma/genetics , Mice , Mice, Knockout , Promoter Regions, Genetic/genetics , Proteasome Endopeptidase Complex/metabolism , STAT4 Transcription Factor/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics
7.
Inflamm Res ; 55(5): 200-6, 2006 May.
Article in English | MEDLINE | ID: mdl-16830107

ABSTRACT

OBJECTIVE AND DESIGN: This study examined the role of neutrophil-derived reactive oxygen species (ROS) in neutrophil recruitment into ultraviolet B (UVB)-exposed skin of mice. METHODS: Mouse dorsal skin was irradiated with UVB (600 mJ/cm2). Accumulation of neutrophils within the inflammatory sites was observed histochemically. Keratinocyte-derived chemokine (KC) and macrophage inflammatory protein 2 (MIP-2) were quantified, and in vivo chemotaxis of neutrophils toward KC and MIP-2 was examined. RESULTS: UVB exposure of mice deficient in myeloperoxidase (MPO), NADPH oxidase, or both, caused skin neutrophil infiltration peaking at 60, 48, and 48 h, respectively, which was earlier than the 72-h peak in wild-type mice. MIP-2 level was higher in mutant than wild-type mice. Mutant neutrophils produced more MIP-2 in vitro. Neutrophil migration toward a localized source of KC was higher in mutant than wild type mice. NADPH oxidase deficiency had a greater effect on migration than MPO deficiency. CONCLUSIONS: These results suggest that ROS produced by neutrophils regulate expression of MIP-2 and migration of neutrophils toward KC. This may explain the earlier infiltration of mutant neutrophils in response to UVB.


Subject(s)
Dermatitis/immunology , NADPH Oxidases/deficiency , Neutrophils/radiation effects , Peroxidase/deficiency , Ultraviolet Rays , Animals , Chemokine CXCL1 , Chemokine CXCL2 , Chemokines/pharmacology , Chemokines, CXC , Chemotaxis, Leukocyte , Dermatitis/metabolism , Dermatitis/pathology , Female , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monokines/pharmacology , NADPH Oxidases/genetics , Neutrophils/immunology , Neutrophils/metabolism , Peroxidase/genetics , Skin/immunology , Skin/pathology , Skin/radiation effects
8.
Biochem Biophys Res Commun ; 345(4): 1504-12, 2006 Jul 14.
Article in English | MEDLINE | ID: mdl-16737685

ABSTRACT

Chemokines are believed to be involved in the pathogenesis of chronic renal failure (CRF). In CRF, significantly increased CCL15-IR plasma concentrations were detected. Whereas in plasma of healthy individuals one predominant CCL15-IR molecule with a M(w) of 15kDa [high molecular weight (HMW-CCL15-IR)] was identified, CRF plasma contains increased concentrations of truncated CCL15-IR molecules [intermediate molecular weight (IMW-CCL15-IR)]. HMW-CCL15-IR isolated from hemofiltrate revealed an M(w) of 10141.3, corresponding to deglycosylated CCL15(1-92) carrying a N-terminal pyrrolidone carboxylic acid. CCL15(12-92) was identified as a major component of IMW-CCL15-IR in CRF plasma. Compared to CCL15(1-92), in monocytes CCL15(12-92) causes stronger induction of intracellular calcium flux, chemotactic activity, and adhesion to fibronectin. Intracellular calcium flux assays revealed that, in comparison to peripheral blood mononuclear cells (PBMC) of healthy donors, PBMCs of CRF patients demonstrated an increased sensitivity to CCL15. Our results point to an involvement of the CCL15-CCR1 axis in the pathophysiology of CRF.


Subject(s)
Chemokines, CC/blood , Kidney Failure, Chronic/blood , Monokines/blood , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Calcium/metabolism , Chemokines, CC/chemistry , Chemokines, CC/pharmacology , Chemotaxis/drug effects , Dose-Response Relationship, Drug , Female , Hemofiltration , Humans , Kidney Failure, Chronic/therapy , Macrophage Inflammatory Proteins , Male , Middle Aged , Molecular Weight , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Monokines/chemistry , Monokines/pharmacology , Peptide Fragments/pharmacology , Renal Dialysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
Life Sci ; 78(2): 134-9, 2005 Nov 26.
Article in English | MEDLINE | ID: mdl-16112685

ABSTRACT

Inhibitory effects of highly purified vitamin B2 (riboflavin-5'-sodium phosphate, >97%) on the interleukin (IL)-6, macrophage inflammatory protein (MIP)-2 and nitric oxide (NO) in LPS-induced shock mice were evaluated. Vitamin B2 at 20 mg/kg (protective effect on mice mortality induced by LPS), intravenously administered 6 h after LPS injection, significantly decreased the plasma elevated levels of IL-6 and MIP-2 at 9 and 12 h. In addition, vitamin B2 lowered the tissue concentration and the mRNA expression of IL-6 in lung and those of MIP-2 in liver at 9 h. Vitamin B2 also reduced concentration of MIP-2 concentration in lung, and inhibited mRNA expression in kidney, respectively. Vitamin B2 decreased the plasma elevated NO levels in accordance with a reduction in expression of inducible NO synthase (iNOS) both at 21 and 24 h. Accordingly, the reduction in elevated plasma cytokine levels and NO based on the inhibitory effect on local cytokine mRNA expression and iNOS would be responsible for the anti-septic effect of vitamin B2.


Subject(s)
Cytokines/biosynthesis , Inflammation/metabolism , Nitric Oxide/biosynthesis , Riboflavin/pharmacology , Shock, Septic/metabolism , Vitamins/pharmacology , Animals , Blotting, Western , Chemokine CXCL2 , Interleukin-6/pharmacology , Kidney/drug effects , Kidney/metabolism , Lipopolysaccharides/pharmacology , Liver/drug effects , Liver/metabolism , Lung/drug effects , Lung/metabolism , Male , Mice , Mice, Inbred ICR , Monokines/pharmacology , Nitric Oxide Synthase Type II/biosynthesis , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
10.
Exp Cell Res ; 307(2): 342-53, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15894313

ABSTRACT

Adhesion, migration and invasion of endothelial cells are prerequisites for the formation of blood vessels and have to be controlled on a subcellular level. We report that subconfluent human umbilical vein endothelial cells (HUVEC) are able to constitutively form podosomal adhesions that are sites of matrix metalloprotease concentration and matrix degradation. Importantly, incubation of serum-starved cells with VEGF or TNFalpha revealed the dependence of podosomes on cytokine signaling. Podosome formation was also stimulated by addition of monocytes to HUVEC. Microinjection/application of specific inhibitors or active/inactive mutants showed that regulatory pathways include Src kinase and RhoGTPase signaling, N-WASP activation and Arp2/3 complex-dependent actin nucleation. In sum, our data show that HUVEC displaying a migratory phenotype constitutively form f-actin-rich adhesions with podosomal characteristics downstream of cytokine signaling. We propose that HUVEC podosomes play an important role in endothelial cell migration and invasion.


Subject(s)
Cell Surface Extensions/metabolism , Cytokines/physiology , Endothelial Cells/cytology , Signal Transduction/physiology , rho GTP-Binding Proteins/physiology , Actin-Related Protein 2 , Actin-Related Protein 3 , Actins/metabolism , Bacterial Toxins/pharmacology , Blood Platelets/physiology , Cell Adhesion/physiology , Cell Communication/physiology , Cell Count , Cell Surface Extensions/drug effects , Cells, Cultured , Coculture Techniques , Culture Media, Serum-Free/pharmacology , Cytokines/pharmacology , Cytoskeletal Proteins/metabolism , Endothelial Cells/metabolism , Endothelial Cells/physiology , Escherichia coli Proteins/pharmacology , Extracellular Matrix/metabolism , Humans , Matrix Metalloproteinases/metabolism , Microinjections , Microscopy, Fluorescence , Monocytes/physiology , Monokines/pharmacology , Mutation/genetics , Nerve Tissue Proteins/pharmacology , Phosphotyrosine/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Vascular Endothelial Growth Factor A/pharmacology , Wiskott-Aldrich Syndrome Protein, Neuronal , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/genetics , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
11.
Blood ; 105(8): 3011-8, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15604218

ABSTRACT

Monocyte cytokines (ie, monokines) induce natural killer (NK) cells to produce interferon-gamma (IFN-gamma), which is critical for monocyte clearance of infectious pathogens and tumor surveillance. Human CD56bright NK cells produce far more IFN-gamma in response to monokines than do CD56dim NK cells. The kinases and phosphatases involved in regulating IFN-gamma production by monokine-activated NK cells are not clearly identified. SHIP1 is a 5' inositol phosphatase that dephosphorylates the phosphatidylinositol-3 kinase (PI-3K) product PI3,4,5P3. Here, we show that constitutive expression of SHIP1 is distinctly lower in CD56bright NK cells compared with CD56dim NK cells, suggesting it could be an important negative regulator of IFN-gamma production in monokine-activated NK cells. Indeed, overexpression of SHIP1 in CD56bright NK cells followed by monokine activation substantially lowered IFN-gamma production. This effect was not seen when NK cells were infected with a SHIP1 mutant containing an inactive catalytic domain. Finally, NK cells in SHIP1-/- mice produced more IFN-gamma in response to monokines in vivo than did NK cells from wild-type mice. Collectively, these results demonstrate that SHIP1 negatively regulates monokine-induced NK cell IFN-gamma production in vitro and in vivo and provide the first molecular explanation for an important functional distinction observed between CD56bright and CD56dim human NK subsets.


Subject(s)
CD56 Antigen/genetics , Killer Cells, Natural/physiology , Monokines/pharmacology , Phosphoric Monoester Hydrolases/genetics , Animals , Cell Line , Down-Regulation/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Inositol Phosphates/metabolism , Inositol Polyphosphate 5-Phosphatases , Interferon-gamma/genetics , Interferon-gamma/metabolism , Killer Cells, Natural/cytology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Stimulation, Chemical
12.
Exp Cell Res ; 297(1): 197-211, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15194436

ABSTRACT

Intrahippocamal injections of kainic acid (KA) significantly increase the expression of monocyte chemoattractant protein-1 (MCP-1) and macrophage inflammatory protein-2 (MIP-2) in the ipsilateral hippocampus at 2-4 h and 21-45 days post-administration, suggesting the possible involvement of these chemokines in both neurodegenerative and regenerative processes. To examine the possible role of these chemokines on neuronal cell death, hippocampal neurons were incubated with either MCP-1 or MIP-2 in vitro and examined to assess the effects on neuronal cell viability. These treatments resulted in significant neuronal apoptosis that could be abrogated by prior treatment with the caspase-1 inhibitor, Z-VAD-FMK, the caspase-3 inhibitor, Z-DEVD-FMK, the Galphai inhibitor, pertussis toxin, or the MAO-B inhibitor, (-)deprenyl. Furthermore, this chemokine apoptotic effect could also be observed in vivo as intrahippocampal injections of MCP-1 or MIP-2 resulted in the apoptosis of hippocampal neurons, thus supporting a direct role of these chemokines in neuronal death. In contrast, immunohistological analysis of kainic acid lesions on days 21-45 revealed significant expression of MCP-1 and MIP-2 associated with reactive astrocytes and macrophages, respectively, with no apoptotic populations being observed. These results suggested that these chemokines might also mediate distinct biological effects on local microenvironmental cell populations at various stages post truama and during cellular repair. To address this possibility, astrocyte were cultured in the presence or absence of these chemokines and examined by microarray analysis for effects on astrocytes gene expression. A number of genes encoding proteins associated with inflammation, cellular signaling, differentiation, and repair were directly modulated by chemokine treatment. More specifically, the RNA and protein expression of the neurotrophic factor, basic fibroblast growth factor (bFGF), was found to be significantly increased upon culture with MCP-1 and MIP-2. Conditioned media derived from chemokine-stimulated astrocytes also facilitated bFGF-dependent neuronal cell differentiation and promoted survival of H19-7 neurons in vitro, suggesting a possible role for chemokine-activated astrocytes as a source of trophic support. Taken together, these data support possible autocrine and paracrine roles for MCP-1 and MIP-2 in both the "death and life" of hippocampal neurons following CNS injury.


Subject(s)
Chemokine CCL2/metabolism , Encephalitis/metabolism , Monokines/metabolism , Nerve Degeneration/metabolism , Nerve Regeneration/physiology , Neurodegenerative Diseases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Astrocytes/drug effects , Astrocytes/immunology , Astrocytes/metabolism , Cell Line, Transformed , Cell Survival/drug effects , Cell Survival/physiology , Chemokine CCL2/pharmacology , Chemokine CXCL2 , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Encephalitis/immunology , Encephalitis/physiopathology , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Hippocampus/immunology , Hippocampus/metabolism , Hippocampus/physiopathology , Kainic Acid , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Monokines/pharmacology , Nerve Degeneration/chemically induced , Nerve Degeneration/immunology , Nerve Regeneration/drug effects , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/physiopathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neurotoxins , Rats , Rats, Inbred F344 , Up-Regulation/drug effects , Up-Regulation/physiology
13.
J Med Virol ; 73(2): 300-8, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15122808

ABSTRACT

Respiratory syncytial virus (RSV) is a major cause of respiratory tract disease in infants, aged adults, and immunosuppressed patients. The only approved medicines for RSV disease are administration of prophylatic antibodies or treatment with a synthetic nucleoside. Both approaches are expensive and the latter is not without risk and of controversial benefit. The present investigation studied whether pharmaceutical or biologic compounds based upon chemokines might be useful in preventing RSV disease. Of interest was RANTES/CCL5, which inhibits infection by HIV strains that use chemokine receptor (CCR)-5 as co-receptor. Herein, we report that prior or simultaneous treatment of HEp-2 cells with recombinant human CCL5 provides dose-dependent inhibition of infection with RSV. Other recombinant chemokines (MIP-1alpha/CCL3, MIP-1beta/CCL4, MCP-2/CCL8, eotaxin/CCL11, MIP-1delta/CCL15, stromal cell derived factor (SDF)-1alpha/CXCL12) were not inhibitory. The data suggested that CCL5 might inhibit infection by blocking fusion (F) protein-epithelial cell interactions. Infections by mutant RSV strains deleted of small hydrophobic and/or attachment proteins and only expressing F protein in the envelope were inhibited by prior treatment with CCL5 or a biologically inactive N-terminally modified met-CCL5. Inhibition was also observed when virus adsorption and treatment with CCL5 were performed at 4 degrees C. Flow cytometry further revealed that epithelial cells were positive for CCR3, but not CCR1 or CCR5. Thus, novel mimetics of CCL5 may be useful prophylatic agents to prevent respiratory tract disease caused by RSV.


Subject(s)
Chemokines, CC/pharmacology , Respiratory Syncytial Viruses/drug effects , Antiviral Agents/pharmacology , Cell Line, Tumor , Chemokine CCL11 , Chemokine CCL3 , Chemokine CCL4 , Chemokine CCL5 , Chemokine CCL8 , Chemokine CXCL12 , Chemokines, CC/metabolism , Chemokines, CXC/pharmacology , Epithelial Cells/virology , Humans , Macrophage Inflammatory Proteins/pharmacology , Monocyte Chemoattractant Proteins/pharmacology , Monokines/pharmacology , Receptors, CCR1 , Receptors, CCR3 , Receptors, CCR5/analysis , Receptors, Chemokine/analysis , Recombinant Proteins/pharmacology , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/physiology , Viral Fusion Proteins/physiology , Virus Replication/drug effects
14.
Pain ; 108(1-2): 67-75, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15109509

ABSTRACT

Endogenous inhibition of inflammatory pain is mediated by leukocytes that secrete opioid peptides upon exposure to stress (cold water swim stress, CWS) or after local injection of corticotropin releasing factor (CRF). Since in early inflammation few opioid-containing leukocytes are detected and since peripheral opioid-mediated antinociception is low we examined whether antinociception could be augmented by increased recruitment of opioid-containing polymorphonuclear cells (PMN). Rats were intraplantarly (i.pl.) injected with Freund's complete adjuvant (FCA) and with the PMN-recruiting chemokine macrophage inflammatory protein-2 (MIP-2, 1-10 microg; control: saline) for 2 h. Intraplantar leukocytes were quantified by flow cytometry. Paw pressure threshold (PPT) was determined before and after exposure to CWS, i.pl. injection of CRF and opioid peptides. Opioid receptors (OR) were measured by binding studies in dorsal root ganglia (DRG) and by immunohistochemistry in the paw. Our studies showed that (i) MIP-2 injection dose-dependently augmented recruitment of PMN and opioid-containing leukocytes (5-fold increase in cells/paw, P < 0.05), (ii) PPT was not different between groups at baseline and after CWS or CRF (maximum MPE: 20+/-2.3-29+/-7.2%, P < 0.05), (iii) injection of opioid peptides dose-dependently increased the PPT (P < 0.05, maximum MPE: and 18+/-2.6-21+/-3.6%), (iv) MOR (micro OR, MOP) binding sites in the ipsilateral DRG were unchanged (24+/-2-22+/-1.2 fmol/mg protein, P < 0.05, ANOVA) and (v) the number of MOR and DOR (delta OR, DOP) stained nerve fibers in peripheral tissue were unaltered (both P > 0.05, t-test). In summary, antinociception during early inflammation is apparently not limited by the number of opioid-containing leukocytes but by OR availability.


Subject(s)
Inflammation/physiopathology , Leukocytes/immunology , Nociceptors/immunology , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Animals , Chemokine CXCL2 , Chemotaxis/drug effects , Enkephalin, Methionine/pharmacology , Flow Cytometry , Freund's Adjuvant , Ganglia, Spinal/metabolism , Inflammation/chemically induced , Leukocytes/cytology , Male , Monokines/pharmacology , Nerve Endings/immunology , Nerve Endings/metabolism , Nociceptors/metabolism , Rats , Rats, Wistar , beta-Endorphin/pharmacology
15.
Arch Virol ; 148(5): 871-90, 2003 May.
Article in English | MEDLINE | ID: mdl-12721796

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV)/ Human herpesvirus 8 encodes three chemokines, which are called viral macrophage inflammatory protein (vMIP)-I, -II, and -III. Here, we expressed the KSHV vMIP-I and vMIP-II proteins and analyzed their biological functions. Both vMIP-I and vMIP-II had an apparent molecular mass of 7.8 kDa and were localized to the cytoplasm in a body cavity-based lymphoma cell line BC-3, stimulated with phorbol ester. We next treated a human monocytic leukemia cell line, THP-1, with purified recombinant vMIP-I and vMIP-II, or vMIP-I and vMIP-II fused with alkaline phosphatase to study Ca(2+) signalling and in vitro chemotaxis in response to these proteins. Calcium mobilization was induced by both vMIP-I and vMIP-II. Furthermore, vMIP-I and vMIP-II induced Ca(2+) mobilization in K562 cells expressing the CC chemokine receptor 5 (CCR5), suggesting that both may be agonistic for CCR5. Additionally, vMIP-I induced Ca(2+) mobilization through the intermediary of CCR8. These viral MIPs were also capable of chemotactically activating the THP-1 cells. These results imply that vMIP-I and vMIP-II may play important roles in the propagation of KS and primary effusion lymphoma by inducing the chemotaxis of CCR5-expressing monocytes.


Subject(s)
Chemotaxis , Herpesvirus 8, Human/physiology , Macrophage Inflammatory Proteins/metabolism , Monocytes/cytology , Monocytes/metabolism , Monokines/metabolism , Signal Transduction , Calcium/metabolism , Calcium Signaling/drug effects , Chemokine CCL1 , Chemokine CCL11 , Chemokine CCL4 , Chemokine CXCL2 , Chemokines, CC/pharmacology , Chemotaxis/drug effects , Dose-Response Relationship, Drug , Gene Expression , Humans , K562 Cells , Macrophage Inflammatory Proteins/genetics , Macrophage Inflammatory Proteins/pharmacology , Monocytes/drug effects , Monokines/genetics , Monokines/pharmacology , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Sarcoma, Kaposi/virology , Signal Transduction/drug effects , Tumor Cells, Cultured
16.
Arthritis Rheum ; 48(3): 668-74, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12632419

ABSTRACT

OBJECTIVE: Adenosine deaminase (ADA; EC 3.5.4.4) activity is elevated in the synovial fluid (SF) of patients with rheumatoid arthritis (RA). Since the antiinflammatory effect of methotrexate is reportedly associated with increased levels of extracellular adenosine, the present study was undertaken to clarify the role of 2 ADA isozymes, ADA1 and ADA2, in the pathogenesis of RA. METHODS: The activities of ADA1 and ADA2 were measured in SF from RA and osteoarthritis (OA) patients, in sera from RA patients, and in lysates prepared from mononuclear and polymorphonuclear cells from SF from RA patients, peripheral blood from RA patients, and fibroblast-like synoviocytes (FLS) from RA and OA patients. Also measured were the effects of proinflammatory cytokines on ADA1 activity and ADA messenger RNA (mRNA) expression in RA FLS, as determined using real-time polymerase chain reaction. The adenosine concentration in RA SF was measured by radioimmunoassay. RESULTS: The adenosine concentration in RA SF ranged from 0.027 microM to 0.508 microM (mean +/- SD 0.156 +/- 0.132 microM). At those concentrations, ADA1 would be expected to be functionally dominant due to its higher affinity for adenosine. ADA1 activity in RA SF (mean +/- SD 14.4 +/- 8.5 IU/liter) was significantly higher than that in OA SF (3.0 +/- 1.1 IU/liter) or RA sera (3.0 +/- 0.6 IU/liter); moreover, ADA1 activity in RA FLS lysate was the highest among the cell lysates tested. Proinflammatory cytokines did not affect ADA1 activity or ADA mRNA expression in RA FLS. CONCLUSION: Elevated ADA1 activity is an intrinsic characteristic of RA FLS, which likely contributes to the pathogenesis of RA by neutralizing the antirheumatic properties of endogenous adenosine.


Subject(s)
Adenosine Deaminase/biosynthesis , Arthritis, Rheumatoid/enzymology , Fibroblasts/enzymology , Osteoarthritis/enzymology , Synovial Fluid/enzymology , Adenosine/metabolism , Adenosine Deaminase/blood , Adenosine Deaminase/genetics , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/pathology , Cells, Cultured , Fibroblasts/cytology , Humans , Isoenzymes , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/enzymology , Monokines/pharmacology , Neutrophils/cytology , Neutrophils/enzymology , Osteoarthritis/pathology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Synovial Fluid/cytology
17.
J Neurochem ; 84(5): 997-1005, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12603824

ABSTRACT

Chemokines are important mediators in immune responses and inflammatory processes of neuroimmunologic and infectious diseases. Although chemokines are expressed predominantly by cells of the immune system, neurons also express chemokines and chemokine receptors. We report herein that human neuronal cells (NT2-N) produce macrophage inflammatory protein-1alpha and -1beta (MIP-1alpha and MIP-1beta), which could be enhanced by interleukin (IL)-1beta at both mRNA and protein levels. The addition of supernatants from human peripheral blood monocyte-derived macrophage (MDM) cultures induced MIP-1beta mRNA expression in NT2-N cells. Anti-IL-1beta antibody removed most, but not all, of the MDM culture supernatant-induced MIP-1beta mRNA expression in NT2-N cells, suggesting that IL-1beta in the MDM culture supernatants is a major factor in the induction of MIP-1beta expression. Investigation of the mechanism(s) responsible for IL-1beta-induced MIP-1alpha and -1beta expression demonstrated that IL-1beta activated nuclear factor kappa B (NF-kappaB) promoter-directed luciferase activity in NT2-N cells. Caffeic acid phenethyl ester, a potent and specific inhibitor of activation of NF-kappaB, not only blocked IL-1beta-induced activation of the NF-kappaB promoter but also decreased IL-1beta-induced MIP-1alpha and -1beta expression in NT2-N cells. These data suggest that NF-kappaB is at least partially involved in the IL-1beta-mediated action on MIP-1alpha and -1beta in NT2-N cells. IL-1beta-mediated up-regulation of beta-chemokine expression may have important implications in the immunopathogenesis of inflammatory diseases in the CNS.


Subject(s)
Interleukin-1/pharmacology , Macrophage Inflammatory Proteins/biosynthesis , Neurons/drug effects , Neurons/metabolism , Cell Line , Chemokine CCL3 , Chemokine CCL4 , Culture Media, Conditioned/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Macrophages/metabolism , Monokines/biosynthesis , Monokines/pharmacology , NF-kappa B/metabolism , Neurons/cytology , Protein Isoforms/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology
18.
Immunol Lett ; 85(1): 1-4, 2003 Jan 02.
Article in English | MEDLINE | ID: mdl-12505189

ABSTRACT

Intracisternal injection of the CXC-chemokines KC or macrophage inflammatory protein (MIP)-2 induced a pleocytosis in the cerebrospinal fluid (CSF) of rats in a dose dependent way. MIP-2 was much more potent than KC. The concurrent injection of both chemokines revealed a profound synergistic effect on leukocyte recruitment into CSF.


Subject(s)
Central Nervous System/physiology , Chemokines, CXC/cerebrospinal fluid , Chemokines, CXC/pharmacology , Chemotactic Factors/pharmacology , Monokines/pharmacology , Neutrophils/drug effects , Animals , Brain/cytology , Chemokine CXCL2 , Dose-Response Relationship, Immunologic , Drug Synergism , Male , Neutrophils/physiology , Rats , Rats, Wistar
19.
J Hepatol ; 36(6): 766-73, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12044526

ABSTRACT

BACKGROUND/AIMS: Recently, we have reported that macrophage inflammatory protein-2 (MIP-2) plays a pivotal role in concanavalin A (Con A)-induced liver injury. In this study, we investigated the effect of antithrombin III (AT-III) on liver damage, and production of MIP-2 and prostacyclin in this model. METHODS: Liver injury was induced by intravenous injection of Con A (15 mg/kg) and AT-III was administered (50, 250 and 500 units/kg, iv) 30 mm before Con A injection. Plasma levels of alanine aminotransferase (ALT), MIP-2 and 6-keto-prostaglandin F1alpha (6k-PG-F1alpha), stable metabolite of prostaglandin I(2) (prostacyclin), were determined. RESULTS: The elevated plasma ALT levels 8, 16, 24 h after Con A injection were inhibited by AT-III pretreatment. The elevated plasma MIP-2 levels were significantly inhibited by AT-III pretreatment compared with vehicle treatment. The inhibitory effect of AT-III on plasma ALT and MIP-2 in Con A-induced liver injury was attenuated by indomethacin (5 mg/kg, ip). Plasma concentration of 6k-PG-F1alpha at 2 h after AT-III injection was significantly elevated compared with baseline and vehicle pretreatment. CONCLUSIONS: These findings suggest that AT-III prevents Con A-induced liver injury through an inhibition of MIP-2 release and a production of prostacyclin.


Subject(s)
Antithrombin III/pharmacology , Chemokines/metabolism , Epoprostenol/biosynthesis , Liver Diseases/drug therapy , Liver Diseases/immunology , Serine Proteinase Inhibitors/pharmacology , Alanine Transaminase/blood , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/pharmacology , Chemical and Drug Induced Liver Injury , Chemokine CXCL2 , Concanavalin A , Cytokines/biosynthesis , Epoprostenol/blood , Female , Indomethacin/pharmacology , Mice , Mice, Inbred BALB C , Monokines/pharmacology , Neutrophils/immunology , Recombinant Proteins/pharmacology , Specific Pathogen-Free Organisms , Tumor Necrosis Factor-alpha/pharmacology
20.
Microvasc Res ; 60(3): 249-60, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11078641

ABSTRACT

The leukocyte integrins LFA-1 and Mac-1 bind to endothelial intercellular adhesion molecule-1 (ICAM-1). Leukocyte adhesion induced by micropipette injection of formylmethionylleucylphenylalanine (fMLP) or macrophage inflammatory protein 2 (MIP-2) next to a venule in the exteriorized mouse cremaster muscle was almost completely blocked after intravenous injection of the ICAM-1 mAb YN-1. In contrast, after 2-h pretreatment with TNF-alpha, leukocyte adhesion induced in postcapillary venules by fMLP or MIP-2 was not blocked by the ICAM-1 mAb. Leukocyte adhesion was significantly reduced by mAb GAME-46 to CD18 even after TNF-alpha treatment. We conclude that ICAM-1 is necessary for neutrophil adhesion to unstimulated endothelium, but not for adhesion to cytokine-stimulated endothelium. Although ICAM-1 is expressed at high levels after TNF-alpha, ICAM-1 either is not functional or is redundant with other endothelial ligands for beta(2) integrins.


Subject(s)
Cell Adhesion/physiology , Intercellular Adhesion Molecule-1/physiology , Leukocytes/physiology , Animals , Antibodies, Monoclonal/pharmacology , CD18 Antigens/physiology , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Movement/physiology , Chemokine CXCL2 , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Inflammation/pathology , Inflammation/physiopathology , Intercellular Adhesion Molecule-1/immunology , Leukocytes/drug effects , Male , Mice , Mice, Inbred C57BL , Monokines/pharmacology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Venules/cytology , Venules/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...