Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
J Immunol ; 206(1): 181-192, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33277387

ABSTRACT

MCP-1-induced monocyte chemotaxis is a crucial event in inflammation and atherogenesis. Identifying the important signal transduction pathways that control monocyte chemotaxis can unravel potential targets for preventive therapies in inflammatory disease conditions. Previous studies have shown that the focal adhesion kinase Pyk2 plays a critical role in monocyte motility. In this study, we investigated the MCP-1-mediated activation of Pyk2 (particularly by the phosphorylation of Tyr402) in primary human peripheral blood monocytes. We showed that MCP-1 induces Src phosphorylation in a similar time frame and that the MCP-1-induced Pyk2 tyrosine phosphorylation is controlled by the Src family kinase. We also report, in this study, that PKCß, an isoform of PKC, is required for both Src and Pyk2 activation/phosphorylation in response to MCP-1 stimulation. We identified Lyn as the specific Src kinase isoform that is activated by MCP-1 and acts upstream of Pyk2 in primary monocytes. Furthermore, Lyn is found to be indispensable for monocyte migration in response to MCP-1 stimulation. Moreover, our coimmunoprecipitation studies in monocytes revealed that PKCß, Pyk2, and Lyn exist constitutively in a molecular complex. To our knowledge, our study has uncovered a novel PKCß-Lyn-Pyk2 signaling cascade in primary monocytes that regulates MCP-1-induced monocyte adhesion and migration.


Subject(s)
Chemokine CCL2/metabolism , Focal Adhesion Kinase 2/metabolism , Monocytes/physiology , Multiprotein Complexes/metabolism , Protein Kinase C beta/metabolism , src-Family Kinases/metabolism , Cell Adhesion , Cells, Cultured , Chemokine CCL2/genetics , Chemotaxis , Humans , Phosphorylation , Primary Cell Culture , Proto-Oncogene Proteins pp60(c-src)/metabolism , Signal Transduction
2.
J Biol Chem ; 293(36): 14040-14064, 2018 09 07.
Article in English | MEDLINE | ID: mdl-30021838

ABSTRACT

Monoamine oxidase A (MAO-A) is a mitochondrial flavoenzyme implicated in the pathogenesis of atherosclerosis and inflammation and also in many neurological disorders. MAO-A also has been reported as a potential therapeutic target in prostate cancer. However, the regulatory mechanisms controlling cytokine-induced MAO-A expression in immune or cancer cells remain to be identified. Here, we show that MAO-A expression is co-induced with 15-lipoxygenase (15-LO) in interleukin 13 (IL-13)-activated primary human monocytes and A549 non-small cell lung carcinoma cells. We present evidence that MAO-A gene expression and activity are regulated by signal transducer and activator of transcription 1, 3, and 6 (STAT1, STAT3, and STAT6), early growth response 1 (EGR1), and cAMP-responsive element-binding protein (CREB), the same transcription factors that control IL-13-dependent 15-LO expression. We further established that in both primary monocytes and in A549 cells, IL-13-stimulated MAO-A expression, activity, and function are directly governed by 15-LO. In contrast, IL-13-driven expression and activity of MAO-A was 15-LO-independent in U937 promonocytic cells. Furthermore, we demonstrate that the 15-LO-dependent transcriptional regulation of MAO-A in response to IL-13 stimulation in monocytes and in A549 cells is mediated by peroxisome proliferator-activated receptor γ (PPARγ) and that signal transducer and activator of transcription 6 (STAT6) plays a crucial role in facilitating the transcriptional activity of PPARγ. We further report that the IL-13-STAT6-15-LO-PPARγ axis is critical for MAO-A expression, activity, and function, including migration and reactive oxygen species generation. Altogether, these results have major implications for the resolution of inflammation and indicate that MAO-A may promote metastatic potential in lung cancer cells.


Subject(s)
Interleukin-13/physiology , Monoamine Oxidase/metabolism , Monocytes/metabolism , A549 Cells , Arachidonate 15-Lipoxygenase/metabolism , Cell Line, Tumor , Cells, Cultured , Humans , Inflammation , Lung Neoplasms/pathology , Monoamine Oxidase/physiology , Neoplasm Metastasis , PPAR gamma/metabolism , STAT6 Transcription Factor/metabolism , U937 Cells
3.
J Immunol ; 198(12): 4855-4867, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28500072

ABSTRACT

Macrophage accumulation is a critical step during development of chronic inflammation, initiating progression of many devastating diseases. Leukocyte-specific integrin αDß2 (CD11d/CD18) is dramatically upregulated on macrophages at inflammatory sites. Previously we found that CD11d overexpression on cell surfaces inhibits in vitro cell migration due to excessive adhesion. In this study, we have investigated how inflammation-mediated CD11d upregulation contributes to macrophage retention at inflammatory sites during atherogenesis. Atherosclerosis was evaluated in CD11d-/-/ApoE-/- mice after 16 wk on a Western diet. CD11d deficiency led to a marked reduction in lipid deposition in aortas and isolated macrophages. Macrophage numbers in aortic sinuses of CD11d-/- mice were reduced without affecting their apoptosis and proliferation. Adoptive transfer of fluorescently labeled wild-type and CD11d-/- monocytes into ApoE-/- mice demonstrated similar recruitment from circulation, but reduced accumulation of CD11d-/- macrophages within the aortas. Furthermore, CD11d expression was significantly upregulated on macrophages in atherosclerotic lesions and M1 macrophages in vitro. Interestingly, expression of the related ligand-sharing integrin CD11b was not altered. This difference defines their distinct roles in the regulation of macrophage migration. CD11d-deficient M1 macrophages demonstrated improved migration in a three-dimensional fibrin matrix and during resolution of peritoneal inflammation, whereas migration of CD11b-/- M1 macrophages was not affected. These results prove the contribution of high densities of CD11d to macrophage arrest during atherogenesis. Because high expression of CD11d was detected in several inflammation-dependent diseases, we suggest that CD11d/CD18 upregulation on proinflammatory macrophages may represent a common mechanism for macrophage retention at inflammatory sites, thereby promoting chronic inflammation and disease development.


Subject(s)
Atherosclerosis/immunology , Blood Vessels/pathology , CD11 Antigens/genetics , CD18 Antigens/genetics , Integrin alpha Chains/genetics , Macrophages/immunology , Animals , Aorta/immunology , Aorta/pathology , Apolipoproteins E/deficiency , Atherosclerosis/etiology , Atherosclerosis/pathology , Blood Vessels/immunology , CD11 Antigens/immunology , CD18 Antigens/immunology , Diet, Western , Humans , Inflammation/pathology , Inflammation Mediators/metabolism , Integrin alpha Chains/deficiency , Integrin alpha Chains/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Peritonitis/immunology , Peritonitis/pathology , Transcriptional Activation , Up-Regulation
4.
J Invest Dermatol ; 135(2): 389-399, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25184961

ABSTRACT

Injury initiates recruitment of macrophages to support tissue repair; however, excessive macrophage activity may exacerbate tissue damage causing further destruction and subsequent delay in wound repair. Here we show that the peroxisome proliferation-activated receptor-γ agonist, rosiglitazone (Rosi), a medication recently reintroduced as a drug to treat diabetes and with known anti-inflammatory properties, paradoxically generates pro-inflammatory macrophages. This is observed in both IL-6-deficient mice and control wild-type mice experimentally induced to produce high titers of auto-antibodies against IL-6, mimicking IL-6 deficiency in human diseases. IL-6 deficiency when combined with Rosi-mediated upregulation of suppressor of cytokine signaling 3 leads to an altered ratio of nuclear signal transducer and activator of transcription 3/NF-κB that allows hyper-induction of inducible nitric oxide synthase (iNOS). Macrophages activated in this manner cause de novo tissue destruction, recapitulating human chronic wounds, and can be reversed in vivo by recombinant IL-6, blocking macrophage infiltration, or neutralizing iNOS. This study provides insight into an unanticipated paradoxical role of Rosi in mediating hyper-inflammatory macrophage activation significant for diseases associated with IL-6 deficiency.


Subject(s)
Inflammation/etiology , Interleukin-6/deficiency , Macrophage Activation/drug effects , Skin/pathology , Thiazolidinediones/pharmacology , Animals , Female , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , NF-kappa B/physiology , Nitric Oxide Synthase Type II/physiology , Rosiglitazone , STAT3 Transcription Factor/physiology , Wound Healing
5.
Article in English | MEDLINE | ID: mdl-26052543

ABSTRACT

Monocytes/macrophages are versatile cells centrally involved in host defense and immunity. Th1 cytokines induce a classical activation program in monocytes/macrophages leading to a proinflammatory M1 macrophage phenotype while Th2 cytokines IL-4 and IL-13 promote monocyte differentiation into an alternatively activated, anti-inflammatory M2 macrophage phenotype. Although monoamine oxidase A (MAO-A) is primarily known for its action in the nervous system, several recent studies have identified MAO-A as a signature marker of alternative activation of monocytes/macrophages. In this brief review we explore the signaling pathways/molecules that regulate MAO-A expression in alternatively activated monocytes/macrophages. We further discuss the contribution of MAO-A to the resolution of inflammation and identify potential therapeutic targets for controlling inflammation. Altogether this review provides deeper insight into the role of MAO-A in alternative activation of monocytes/macrophages and their participation in the inflammatory response.

6.
Inflamm Res ; 62(11): 991-1001, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23974215

ABSTRACT

OBJECTIVE AND DESIGN: We designed a study to detect downstream phosphorylation targets of PKCß in MCP-1-induced human monocytes. METHODS: Two-dimensional gel electrophoresis was performed for monocytes treated with MCP-1 in the presence or absence of PKCß antisense oligodeoxyribonucleotides (AS-ODN) or a PKCß inhibitor peptide, followed by phospho- and total protein staining. Proteins that stained less intensely with the phospho-stain, when normalized to the total protein stain, in the presence of PKCß AS-ODN or the PKCß inhibitor peptide, were sequenced. RESULTS: Of the proteins identified, vimentin was consistently identified using both experimental approaches. Upon (32)P-labeling and vimentin immunoprecipitation, increased phosphorylation of vimentin was observed in MCP-1 treated monocytes as compared to the untreated monocytes. Both PKCß AS-ODN and the PKCß inhibitor reduced MCP-1-induced vimentin phosphorylation. The IP of monocytes with anti-vimentin antibody and immunoblotting with a PKCß antibody revealed that increased PKCß becomes associated with vimentin upon MCP-1 activation. Upon MCP-1 treatment, monocytes were shown to secrete vimentin and secretion depended on PKCß expression and activity. CONCLUSIONS: We conclude that vimentin, a major intermediate filament protein, is a phosphorylation target of PKCß in MCP-1-treated monocytes and that PKCß phosphorylation is essential for vimentin secretion. Our recently published studies have implicated vimentin as a potent stimulator of the innate immune receptor Dectin-1 as reported by Thiagarajan et al. (Cardiovasc Res 99:494-504, 2013). Taken together our findings suggest that inhibition of PKCß regulates vimentin secretion and, thereby, its interaction with Dectin-1 and downstream stimulation of superoxide anion production. Thus, PKCß phosphorylation of vimentin likely plays an important role in propagating inflammatory responses.


Subject(s)
Chemokine CCL2/immunology , Monocytes/immunology , Protein Kinase C beta/immunology , Vimentin/immunology , Cells, Cultured , Humans , Phosphorylation
7.
Cardiovasc Res ; 99(3): 494-504, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23674515

ABSTRACT

AIMS: Atherosclerosis is a chronic inflammatory disorder of cholesterol deposition in monocyte-derived macrophages (MDM) within the arterial wall leading to impingement on the lumen of the vessel. In atherosclerotic lesions, MDM are the primary source of NADPH oxidase-derived superoxide anion (O2⁻) inducing low-density lipoprotein (LDL) oxidation leading to their unregulated uptake of oxidized LDL and foam cell formation. We recently discovered that zymosan potently activates monocyte NADPH oxidase via the non-toll pattern recognition receptor (PRR), Dectin-1. Other PRRs bind endogenous human ligands, yet no such ligands have been identified for Dectin-1. Our hypothesis was that inflammation generates endogenous ligands for Dectin-1 that activate O2⁻ production and thereby contributes to atherogenesis. METHODS AND RESULTS: Human: anti-zymosan antibodies were used to identify similar, cross-reactive epitopes in human atherosclerotic tissue extracts. Immunoblot analysis revealed consistent antibody reactive protein bands on one- and two-dimensional gel electrophoreses. Vimentin was identified by mass spectrometry in the immunoreactive bands across different tissue samples. Direct binding of vimentin to Dectin-1 was observed using BIACORE. Further data revealed that vimentin induces O2⁻ production by human monocytes. Analysis of human atherosclerotic lesions revealed that vimentin was detected extracellularly in the necrotic core and in areas of active inflammation. Vimentin also co-localized with Dectin-1 in macrophage-rich regions where O2⁻ is produced. CONCLUSION: We conclude that vimentin is an endogenous, activating ligand for Dectin-1. Its presence in areas of artery wall inflammation and O2⁻ production suggests that vimentin activates Dectin-1 and contributes to the oxidation of lipids and cholesterol accumulation in atherosclerosis.


Subject(s)
Carotid Stenosis/immunology , Carotid Stenosis/metabolism , Lectins, C-Type/metabolism , Vimentin/metabolism , Carotid Stenosis/pathology , Cholesterol/metabolism , Electrophoresis, Gel, Two-Dimensional , Humans , Immunoblotting , Ligands , Lipid Metabolism , Macrophages/metabolism , Oxidation-Reduction , Protein Binding , Receptors, Pattern Recognition/metabolism , Superoxides/metabolism , Zymosan/metabolism
8.
Free Radic Biol Med ; 54: 1-16, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23124025

ABSTRACT

Monocytes/macrophages are innate immune cells that play a crucial role in the resolution of inflammation. In the presence of the Th2 cytokines interleukin-4 (IL-4) and interleukin-13 (IL-13), they display an anti-inflammatory profile and this activation pathway is known as alternative activation. In this study we compare and differentiate pathways mediated by IL-4 and IL-13 activation of human monocytes/macrophages. Here we report differential regulation of IL-4 and IL-13 signaling in monocytes/macrophages starting from IL-4/IL-13 cytokine receptors to Jak/Stat-mediated signaling pathways that ultimately control expression of several inflammatory genes. Our data demonstrate that although the receptor-associated tyrosine kinases Jak2 and Tyk2 are activated after the recruitment of IL-13 to its receptor (containing IL-4Rα and IL-13Rα1), IL-4 stimulates Jak1 activation. We further show that Jak2 is upstream of Stat3 activation and Tyk2 controls Stat1 and Stat6 activation in response to IL-13 stimulation. In contrast, Jak1 regulates Stat3 and Stat6 activation in IL-4-induced monocytes. Our results further reveal that although IL-13 utilizes both IL-4Rα/Jak2/Stat3 and IL-13Rα1/Tyk2/Stat1/Stat6 signaling pathways, IL-4 can use only the IL-4Rα/Jak1/Stat3/Stat6 cascade to regulate the expression of some critical inflammatory genes, including 15-lipoxygenase, monoamine oxidase A (MAO-A), and the scavenger receptor CD36. Moreover, we demonstrate here that IL-13 and IL-4 can uniquely affect the expression of particular genes such as dual-specificity phosphatase 1 and tissue inhibitor of metalloprotease-3 and do so through different Jaks. As evidence of differential regulation of gene function by IL-4 and IL-13, we further report that MAO-A-mediated reactive oxygen species generation is influenced by different Jaks. Collectively, these results have major implications for understanding the mechanism and function of alternatively activated monocytes/macrophages by IL-4 and IL-13 and add novel insights into the pathogenesis and potential treatment of various inflammatory diseases.


Subject(s)
Interleukin-13/immunology , Interleukin-4/immunology , Macrophages/immunology , Monocytes/immunology , Arachidonate 15-Lipoxygenase/metabolism , Cells, Cultured , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 1/metabolism , Gene Expression Regulation , Humans , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Macrophage Activation/immunology , Monoamine Oxidase/metabolism , Reactive Oxygen Species/metabolism , Receptors, Interleukin-13/metabolism , Receptors, Interleukin-4/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/immunology , TYK2 Kinase/metabolism , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/metabolism
9.
J Lipid Res ; 54(2): 436-47, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23160182

ABSTRACT

Monocyte chemoattractant protein-1 (MCP-1)-induced monocyte chemotaxis is a major event in inflammatory disease. Our prior studies have demonstrated that MCP-1-dependent chemotaxis requires release of arachidonic acid (AA) by activated cytosolic phospholipase A(2) (cPLA(2)). Here we investigated the involvement of AA metabolites in chemotaxis. Neither cyclooxygenase nor lipoxygenase pathways were required, whereas pharmacologic inhibitors of both the cytochrome-P450 (CYP) and the soluble epoxide hydrolase (sEH) pathways blocked monocyte chemotaxis to MCP-1. To verify specificity, we demonstrated that the CYP and sEH products epoxyeiscosatrienoic acids (EETs) and dihydroxyeicosatrienoic acids (DHETs), respectively, restored chemotaxis in the presence of the inhibitors, indicating that sEH-derived products are essential for MCP-1-driven chemotaxis. Importantly, DHETs also rescued chemotaxis in cPLA(2)-deficient monocytes and monocytes with blocked Erk1/2 activity, because Erk controls cPLA(2) activation. The in vitro findings regarding the involvement of CYP/sEH pathways were further validated in vivo using two complementary approaches measuring MCP-1-dependent chemotaxis in mice. These observations reveal the importance of sEH in MCP-1-regulated monocyte chemotaxis and may explain the observed therapeutic value of sEH inhibitors in treatment of inflammatory diseases, cardiovascular diseases, pain, and even carcinogenesis. Their effectiveness, often attributed to increasing EET levels, is probably influenced by the impairment of DHET formation and inhibition of chemotaxis.


Subject(s)
Chemokine CCL2/metabolism , Chemotaxis , Epoxide Hydrolases/chemistry , Epoxide Hydrolases/metabolism , Monocytes/cytology , Animals , Arachidonic Acid/biosynthesis , Chemotaxis/drug effects , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/antagonists & inhibitors , Fatty Acids, Monounsaturated/chemistry , Fatty Acids, Monounsaturated/metabolism , Female , Humans , Lipoxygenase/metabolism , Mice , Monocytes/drug effects , Monocytes/enzymology , Monocytes/metabolism , Phospholipases A2, Cytosolic/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Solubility
10.
J Biol Chem ; 288(4): 2778-88, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23184931

ABSTRACT

IL-13 is a potent stimulator of alternative monocyte/macrophage activation. During alternative activation, the expression of several proteins is induced including 15-lipoxygenase (15-LO), a lipid-peroxidating enzyme and the scavenger receptor CD36. We previously reported that α(M)ß(2) integrin activation or clustering suppresses the expression of both 15-LO and CD36. In this study we focused on exploring the molecular mechanisms that down-regulate CD36 expression and CD36-mediated foam cell formation in IL-13-stimulated monocytes/macrophages after α(M)ß(2) activation. Our studies reveal that α(M)ß(2) integrin activation inhibits the IL-13 activation of several critical pathways that are required for macrophage alternative activation; namely, blocking Jak2 and Tyk2 phosphorylation, which bind to the cytoplasmic tails of the IL-4Rα/IL-13Rα1 complex. This leads to the inhibition of tyrosine phosphorylation of Stats (Stat1, Stat3, and Stat6) and prevents the formation of a signaling complex (containing p38MAPK, PKCδ, and Stat3) that are critical for the expression of both 15-LO and CD36. Jak2-mediated Hck activation is also inhibited, thereby preventing Stats serine phosphorylation, which is essential for downstream Stat-dependent gene transcription. Moreover, inhibition of Jak2, Tyk2, or their downstream target 15-LO with antisense oligonucleotides profoundly inhibits IL-13-induced CD36 expression and CD36-dependent foam cell formation, whereas13(S) Hydroperoxyoctadecadienoic acid (HPODE), a 15-LO product and peroxisome proliferator-activated receptor-γ ligand, completely restores CD36 expression in monocytes treated with 15-LO antisense. α(M)ß(2) integrin activation controls CD36 expression and foam cell formation in alternatively activated monocyte/macrophages by blocking Tyk2/Jak2 phosphorylation via a 15-LO-dependent pathway. The discovery of this mechanism helps our understanding of the potential role of alternatively activated macrophages in atherogenesis and highlights the impact of integrin α(M)ß(2) on this process.


Subject(s)
Foam Cells/cytology , Macrophage-1 Antigen/metabolism , Macrophages/metabolism , Receptors, Interleukin-13/metabolism , Animals , Atherosclerosis , CD36 Antigens/biosynthesis , Cell Separation , Female , Flow Cytometry , Humans , Interleukin-13/metabolism , Interleukin-4/metabolism , Janus Kinase 2/metabolism , Lipids/chemistry , Macrophages/cytology , Mice , Signal Transduction , TYK2 Kinase/metabolism
13.
J Biol Chem ; 286(42): 36709-23, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-21878628

ABSTRACT

IL-13 is a Th2 cytokine that promotes alternative activation (M2 polarization) in primary human monocytes. Our studies have characterized the functional IL-13 receptor complex and the downstream signaling events in response to IL-13 stimulation in alternatively activated monocytes/macrophages. In this report, we present evidence that IL-13 induces the activation of a Src family tyrosine kinase, which is required for IL-13 induction of M2 gene expression, including 15-lipoxygenase (15-LO). Our data show that Src kinase activity regulates IL-13-induced p38 MAPK tyrosine phosphorylation via the upstream kinases MKK3 or MKK6. Our findings also reveal that the IL-13 receptor-associated tyrosine kinase Jak2 is required for the activation of both Src kinase as well as p38 MAPK. Further, we found that Src tyrosine kinase-mediated activation of p38 MAPK is required for Stat1 and Stat3 serine 727 phosphorylation in alternatively activated monocytes/macrophages. Additional studies identify Hck as the specific Src family member, stimulated by IL-13 and involved in regulating both p38 MAPK activation and p38 MAPK-mediated 15-LO expression. Finally we show that the Hck regulates the expression of other alternative state (M2)-specific genes (Mannose receptor, MAO-A, and CD36) and therefore conclude that Hck acts as a key regulator controlling gene expression in alternatively activated monocytes/macrophages.


Subject(s)
Gene Expression Regulation/physiology , MAP Kinase Signaling System/physiology , Macrophage Activation/physiology , Monocytes/metabolism , Proto-Oncogene Proteins c-hck/metabolism , Arachidonate 15-Lipoxygenase/biosynthesis , CD36 Antigens/metabolism , Enzyme Activation/physiology , Humans , Interleukin-13/biosynthesis , Janus Kinase 2/metabolism , Lectins, C-Type/metabolism , MAP Kinase Kinase 3/metabolism , MAP Kinase Kinase 6/metabolism , Mannose Receptor , Mannose-Binding Lectins/metabolism , Monoamine Oxidase/metabolism , Monocytes/cytology , Phosphorylation/physiology , Receptors, Cell Surface/metabolism , Receptors, Interleukin-13/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , src-Family Kinases/metabolism
14.
J Leukoc Biol ; 90(3): 599-611, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21653233

ABSTRACT

Zymosan, a mimic of fungal pathogens, and its opsonized form (ZOP) are potent stimulators of monocyte NADPH oxidase, resulting in the production of O(2)(.-), which is critical for host defense against fungal and bacterial pathogens and efficient immune responses; however, uncontrolled O(2)(.-) production may contribute to chronic inflammation and tissue injury. Our laboratory has focused on characterizing the signal transduction pathways that regulate NADPH oxidase activity in primary human monocytes. In this study, we examined the involvement of various pattern recognition receptors and found that Dectin-1 is the primary receptor for zymosan stimulation of O(2)(.-) via NADPH oxidase in human monocytes, whereas Dectin-1 and CR3 mediate the activation by ZOP. Further studies identified Syk and Src as important signaling components downstream of Dectin-1 and additionally identified PKCδ as a novel downstream signaling component for zymosan-induced O(2)(.-) as well as phagocytosis. Our results show that Syk and Src association with Dectin-1 is dependent on PKCδ activity and expression and demonstrate direct binding between Dectin-1 and PKCδ. Finally, our data show that PKCδ and Syk but not Src are required for Dectin-1-mediated phagocytosis. Taken together, our data identify Dectin-1 as the major PRR for zymosan in primary human monocytes and identify PKCδ as a novel downstream signaling kinase for Dectin-1-mediated regulation of monocyte NADPH oxidase and zymosan phagocytosis.


Subject(s)
Membrane Proteins/metabolism , Monocytes/metabolism , NADPH Oxidases/metabolism , Nerve Tissue Proteins/metabolism , Protein Kinase C-delta/metabolism , Signal Transduction , Zymosan/metabolism , Blotting, Western , Cells, Cultured , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Lectins, C-Type , Leukocytes/cytology , Leukocytes/metabolism , Macrophage-1 Antigen/metabolism , Monocytes/cytology , Monocytes/drug effects , Phagocytosis , Phosphorylation , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Superoxides/metabolism , Surface Plasmon Resonance , Syk Kinase , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Tyrosine/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
15.
Mol Cancer Ther ; 10(7): 1149-60, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21596889

ABSTRACT

Glioblastoma multiforme (GBM) is one of the most lethal forms of cancer, with a survival rate of only 13% to 27% within 2 years of diagnosis despite optimal medical treatment. We hypothesize that the presence of a unique IL-13Rα2 decoy receptor prevents GBM apoptosis. This receptor has a high affinity for interleukin-13 (IL-13), binds the cytokine, and competitively inhibits the intracellular signaling cascade initiated by IL-13. In cells lacking the IL-13Rα2 decoy receptor, IL-13 initiates the production of 15-lipoxygenase-1 (15-LOX-1), which has been implicated in cellular apoptosis. Our group and others have shown that induction of 15-LOX-1 correlates with tumor cell death in colorectal, pancreatic, and prostate cancer. How 15-LOX-1 induces apoptosis remains unclear. Preliminary evidence in GBM cells implicates an apoptotic process mediated by PPARγ. 15-LOX-1 metabolites can modulate PPARγ and activation of PPARγ can suppress tumor growth. We hypothesize that in GBM, IL-13 can induce 15-LOX-1, which regulates cell apoptosis via signaling through PPARγ and that expression of IL-13Rα2 prevents apoptosis and contributes to tumor growth. Our in vitro and in vivo data support this. Knocking down IL-13Rα2 with short interfering RNA dramatically induces 15-LOX-1 expression, promotes apoptosis, and reduces GBM tumor growth in vivo. These findings identify a mechanism for eliminating the blockade of endogenous IL-13 signaling and for promotion of apoptosis, and characterize a role for 15-LOX-1 in GBM apoptosis. Identifying a mechanistic pathway that can be targeted for pharmacologic intervention will have applied implications to developing novel and effective treatments of GBM.


Subject(s)
Cell Death/genetics , Gene Silencing , Glioblastoma/pathology , Interleukin-13 Receptor alpha2 Subunit/genetics , Interleukin-13 Receptor alpha2 Subunit/metabolism , Signal Transduction/genetics , Animals , Apoptosis/genetics , Arachidonate 15-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Ligands , Mice , Mice, Nude , PPAR gamma/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Tumor Burden/genetics
16.
Circ Res ; 108(5): 544-54, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21252155

ABSTRACT

RATIONALE: The alternative activation of monocytes by interleukin (IL)-13 and IL-4 is a significant component of the inflammatory response. The consequences of alternative activation in inflammatory diseases remain to be determined. OBJECTIVE: In this report, we explored how integrins, receptors important for monocyte migration to inflammatory sites, regulate IL-13-mediated monocyte activation. We focused on the analysis of 2 proteins, which are upregulated during the alternative activation and are important for the development of atherosclerosis, an oxidative enzyme 15-lipoxygenase (15-LO) and a scavenger receptor CD36. METHODS AND RESULTS: We found that adhesion of resting monocytes through ß(2) integrins and inside-out activation of ß(2) integrins by monocyte chemoattractant protein-1 did not change IL-13-stimulated 15-LO upregulation; however, preincubation of monocytes with the antibody MEM48, which generates full activation of ß(2) integrins, significantly inhibited 15-LO mRNA and protein expression. In contrast, activation of ß(1) integrins had no effect on 15-LO expression. Analysis of integrin clustering through α(M), α(L), α(X), and α(D) subunits demonstrated the pivotal role for integrin α(M)ß(2) in inhibiting 15-LO expression. IL-13 treatment upregulates 15-LO-dependent CD36 expression on human monocytes; our studies showed that ß(2) integrin activation and α(M) integrin clustering significantly inhibited IL-13-dependent CD36 mRNA and protein expression, as well as CD36-related foam cell formation. Moreover, IL-13 stimulation of α(M)-deficient peritoneal macrophages demonstrated an upregulated level of 15-LO induction, CD36 expression, and lipid accumulation as compared with wild-type controls. CONCLUSIONS: The adhesion of monocytes/macrophages through activated integrin α(M)ß(2) has a regulatory and potential atheroprotective function during the alternative activation of macrophages.


Subject(s)
Foam Cells/cytology , Foam Cells/metabolism , Macrophage Activation/physiology , Macrophage-1 Antigen/metabolism , Macrophages/cytology , Macrophages/metabolism , Animals , Arachidonate 15-Lipoxygenase/metabolism , CD11b Antigen/genetics , CD11b Antigen/metabolism , CD36 Antigens/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Humans , Interleukin-13/pharmacology , Lectins, C-Type/metabolism , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Mice, Knockout , Models, Animal , Peptide Fragments/metabolism , Receptors, Cell Surface/metabolism , Receptors, IgG/metabolism
17.
J Immunol ; 185(9): 5211-24, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20861348

ABSTRACT

IL-13 induces profound expression of 15-lipoxygenase (15-LO) in primary human monocytes. Our studies have defined the functional IL-13R complex, association of Jaks with the receptor components, and the tyrosine phosphorylation of several Stat molecules in response to IL-13. Furthermore, we identified both p38MAPK and protein kinase Cδ as critical regulators of 15-LO expression. In this study, we report an ERK1/2-dependent signaling cascade that regulates IL-13-mediated 15-LO gene expression. We show the rapid phosphorylation/activation of ERK1/2 upon IL-13 exposure. Our results indicate that Tyk2 kinase is required for the activation of ERK1/2, which is independent of the Jak2, p38MAPK, and protein kinase Cδ pathways, suggesting bifurcating parallel regulatory pathways downstream of the receptor. To investigate the signaling mechanisms associated with the ERK1/2-dependent expression of 15-LO, we explored the involvement of transcription factors, with predicted binding sites in the 15-LO promoter, in this process including Elk1, early growth response-1 (Egr-1), and CREB. Our findings indicate that IL-13 induces Egr-1 nuclear accumulation and CREB serine phosphorylation and that both are markedly attenuated by inhibition of ERK1/2 activity. We further show that ERK1/2 activity is required for both Egr-1 and CREB DNA binding to their cognate sequences identified within the 15-LO promoter. Furthermore, by transfecting monocytes with the decoy oligodeoxyribonucleotides specific for Egr-1 and CREB, we discovered that Egr-1 and CREB are directly involved in regulating 15-LO gene expression. These studies characterize an important regulatory role for ERK1/2 in mediating IL-13-induced monocyte 15-LO expression via the transcription factors Egr-1 and CREB.


Subject(s)
Arachidonate 15-Lipoxygenase/biosynthesis , Gene Expression Regulation/immunology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Monocytes/enzymology , Arachidonate 15-Lipoxygenase/immunology , CREB-Binding Protein , DNA, Single-Stranded , Enzyme Activation/immunology , Enzyme-Linked Immunosorbent Assay , Gene Expression , Humans , Immunoblotting , Immunoprecipitation , Interleukin-13/immunology , Interleukin-13/metabolism , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/immunology , Monocytes/immunology , Phosphorylation , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Transfection
18.
J Lipid Res ; 50 Suppl: S231-6, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19109234

ABSTRACT

Signal-activated phospholipases are a recent focus of the rapidly growing field of lipid signaling. The extent of their impact on the pathways regulating diverse cell functions is beginning to be appreciated. A critical step in inflammation is the attraction of leukocytes to injured or diseased tissue. Chemotaxis of leukocytes, a requisite process for monocyte and neutrophil extravasation from the blood into tissues, is a critical step for initiating and maintaining inflammation in both acute and chronic settings. Recent studies have identified new important and required roles for two signal-activated phospholipases A2 (PLA2) in regulating chemotaxis. The two intracellular phospholipases, cPLA2alpha (Group IVA) and iPLA2beta (Group VIA), act in parallel to provide distinct lipid mediators at different intracellular sites that are both required for leukocytes to migrate toward the chemokine monocyte chemoattractant protein-1. This review will summarize the separate roles of these phospholipases as well as what is currently known about the influence of two other classes of intracellular signal-activated phospholipases, phospholipase C and phospholipase D, in regulating chemotaxis in eukaryotic cells, but particularly in human monocytes. The contributions of these phospholipases to chemotaxis both in vitro and in vivo will be highlighted.


Subject(s)
Chemotaxis, Leukocyte , Phospholipases/metabolism , Signal Transduction , Animals , Humans , Phosphoinositide Phospholipase C/classification , Phosphoinositide Phospholipase C/metabolism , Phospholipase D/classification , Phospholipase D/metabolism , Phospholipases/classification , Phospholipases A2/classification , Phospholipases A2/metabolism
19.
J Immunol Methods ; 330(1-2): 86-95, 2008 Jan 31.
Article in English | MEDLINE | ID: mdl-18191414

ABSTRACT

Identification of novel signal transduction pathways regulating monocyte chemotaxis can indicate unique targets for preventive therapies for treatment of chronic inflammatory diseases. To aid in this endeavor we report conditions for optimal transfection of primary human monocytes coupled with a new model system for assessing their chemotactic activity in vivo. This method can be used as a tool to identify the relevant signal transduction pathways regulating human monocyte chemotaxis to MCP-1 in the complex in vivo environment that were previously identified to regulate chemotaxis in vitro. MCP-1-dependent chemotaxis of monocytes is studied in an adoptive transfer model where human monocytes transfected with mutant cDNAs are transferred to mice followed by initiation of peritonitis. Harvesting peritoneal cells at 24 h diminishes the contribution of immunologic responses to the cross-species transfer. Validation of relevant regulatory molecules in vivo is critical for understanding the most relevant therapeutic targets for drug development.


Subject(s)
Biological Assay/methods , Chemokine CCL2/metabolism , Chemotaxis, Leukocyte , Monocytes/immunology , Peritonitis/immunology , Protein Kinase C/metabolism , Signal Transduction/immunology , Transfection/methods , Adoptive Transfer , Animals , Cell Movement , Cell Survival , Cells, Cultured , Chemotaxis, Leukocyte/genetics , Disease Models, Animal , Feasibility Studies , Green Fluorescent Proteins/metabolism , Humans , Mice , Monocytes/enzymology , Monocytes/transplantation , Mutation , Peritonitis/chemically induced , Peritonitis/enzymology , Protein Kinase C/genetics , Protein Kinase C beta , Recombinant Fusion Proteins/metabolism , Reproducibility of Results , Signal Transduction/genetics , Thioglycolates , Time Factors
20.
J Exp Med ; 205(2): 347-59, 2008 Feb 18.
Article in English | MEDLINE | ID: mdl-18208975

ABSTRACT

Monocyte chemoattractant protein-1 (MCP-1) directs migration of blood monocytes to inflamed tissues. Despite the central role of chemotaxis in immune responses, the regulation of chemotaxis by signal transduction pathways and their in vivo significance remain to be thoroughly deciphered. In this study, we examined the intracellular location and functions of two recently identified regulators of chemotaxis, Ca(2+)-independent phospholipase (iPLA(2)beta) and cytosolic phospholipase (cPLA(2)alpha), and substantiate their in vivo importance. These enzymes are cytoplasmic in unstimulated monocytes. Upon MCP-1 stimulation, iPLA(2)beta is recruited to the membrane-enriched pseudopod. In contrast, cPLA(2)alpha is recruited to the endoplasmic reticulum. Although iPLA(2)beta or cPLA(2)alpha antisense oligodeoxyribonucleotide (ODN)-treated monocytes display reduced speed, iPLA(2)beta also regulates directionality and actin polymerization. iPLA(2)beta or cPLA(2)alpha antisense ODN-treated adoptively transferred mouse monocytes display a profound defect in migration to the peritoneum in vivo. These converging observations reveal that iPLA(2)beta and cPLA(2)alpha regulate monocyte migration from different intracellular locations, with iPLA(2)beta acting as a critical regulator of the cellular compass, and identify them as potential targets for antiinflammatory strategies.


Subject(s)
Chemokine CCL2/immunology , Group IV Phospholipases A2/immunology , Group VI Phospholipases A2/immunology , Monocytes/immunology , Actins/metabolism , Adoptive Transfer , Animals , Arachidonic Acids/pharmacology , Cells, Cultured , Chemotaxis/drug effects , Chemotaxis/immunology , Female , Group IV Phospholipases A2/antagonists & inhibitors , Group VI Phospholipases A2/antagonists & inhibitors , Humans , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred BALB C , Monocytes/drug effects , Naphthalenes/pharmacology , Oligodeoxyribonucleotides, Antisense/pharmacology , Peritonitis/chemically induced , Peritonitis/immunology , Pyrones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...