Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Biomed Mater Res A ; 103(4): 1380-90, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25045023

ABSTRACT

The effects of different material surfaces on phenotypic expression in macrophages and foreign body giant cells (FBGC) were addressed using our in vitro system of interleukin (IL)-4-induced macrophage fusion and FBGC formation. Arginine-glycine-aspartate (RGD)-, vitronectin (VN)-, and chitosan (CH)-adsorbed cell culture polystyrene, carboxylated (C, negatively charged) polystyrene, and unmodified (PS, non-cell culture treated) polystyrene were compared for their abilities to support monocyte/macrophage adhesion and IL-4-induced macrophage fusion. Pooled whole cell lysates from four different donors were evaluated by immunoblotting for expression of selected components in monocytes, macrophages, and FBGC. In addition to RGD and VN as previously shown, we find that CH supports macrophage adhesion and FBGC formation, whereas C or PS support macrophage adhesion but do not permit macrophage fusion under otherwise identical conditions of IL-4 stimulation. Likewise, components related to macrophage fusion (CD206, CD98, CD147, CD13) are strongly expressed on RGD-, VN-, and CH-adsorbed surfaces but are greatly diminished or not detected on C or PS. Importantly, material surfaces also influence the FBGC phenotype itself, as demonstrated by strong differences in patterns of expression of HLA-DR, B7-2, B7-H1, and toll-like receptor (TLR)-2 on RGD, VN, and CH despite morphologic similarities between FBGC on these surfaces. Likewise, we observe differences in the expression of B7-2, α2-macroglobulin, TLR-2, and fascin-1 between mononuclear macrophages on C and PS. Collectively, these findings reveal the extent to which material surface chemistry influences macrophage/FBGC phenotype beyond evident morphological similarities or differences and identify CH as an FBGC-supportive substrate.


Subject(s)
Giant Cells, Foreign-Body/cytology , Interleukin-4/pharmacology , Macrophages/cytology , Materials Testing , Monocytes/cytology , Biomarkers/metabolism , Cell Fusion , Giant Cells, Foreign-Body/drug effects , Humans , Lymphocytes/cytology , Lymphocytes/drug effects , Macrophages/drug effects , Macrophages/metabolism , Phenotype , Surface Properties , Time Factors , Toll-Like Receptors/metabolism
2.
J Biomed Mater Res A ; 102(6): 2017-23, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24616384

ABSTRACT

In previous studies that explored the influence of cytokines on foreign body giant cell (FBGC) formation, we focused on interleukin (IL)-4 and IL-13, each of which was discovered to induce macrophage fusion leading to FBGC formation in vitro. Two correlative in vivo studies also confirmed that IL-4 plays a role in FBGC formation on implanted biomaterials, but that T lymphocytes are not the source of IL-4 or other cytokines that support this process. The present study focused on identification of the cellular source of macrophage fusion-inducing cytokines, including natural killer (NK) or NKT lymphocytes and mast cells using mouse models genetically deficient in each of these cell types, as well as IL-4 receptor alpha(IL-4Rα)-deficient and severe combined immunodeficient (SCID) mice. Polyetherurethane (PEU) and polyethylene terephthalate (PET) polymers were subcutaneously implanted and retrieved after 14, 21, or 28 days. FBGC formation was evaluated using quantitative and qualitative data from retrieved polymer surfaces. Both types of data indicate that, compared to normal control mice, neither NK or NKT lymphocytes nor mast cells are required for FBGC formation. Furthermore, FBGC formation on biomaterials can proceed in IL-4Rα-deficient and in SCID mice. Similar conclusions were made regarding FBGC formation on both PEU and PET biomaterials. These data suggest that other sources of IL-4/IL-13 and/or additional macrophage fusion-inducing cytokines can mediate FBGC formation on implanted biomaterials, or that, in the absence of normal primary pathways, FBGC formation is nevertheless supported by redundant innate mechanisms.


Subject(s)
Biocompatible Materials/chemistry , Giant Cells, Foreign-Body/cytology , Interleukin-4 Receptor alpha Subunit/genetics , Killer Cells, Natural/immunology , Mast Cells/immunology , Polyethylene Terephthalates/chemistry , Polyurethanes/chemistry , Animals , Female , Gene Deletion , Giant Cells, Foreign-Body/immunology , Interleukin-4/immunology , Interleukin-4 Receptor alpha Subunit/immunology , Mice , Mice, SCID , Prostheses and Implants
3.
Tissue Eng Part A ; 20(1-2): 250-63, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23937279

ABSTRACT

Macrophages are phagocytic cells with great importance in guiding multiple stages of inflammation and tissue repair. By producing a large number of biologically active molecules, they can affect the behavior of other cells and events, such as the foreign body response and angiogenesis. Since protein adsorption to biomaterials is crucial for the inflammatory process, we addressed the ability of the pro-inflammatory molecule fibrinogen (Fg) to modulate macrophage behavior toward tissue repair/regeneration. For this purpose, we used chitosan (Ch) as a substrate for Fg adsorption. Freshly isolated human monocytes were seeded on Ch substrates alone or previously adsorbed with Fg, and allowed to differentiate into macrophages for 10 days. Cell adhesion and morphology, formation of foreign body giant cells (FBGC), and secretion of a total of 80 cytokines and growth factors were evaluated. Both substrates showed similar numbers of adherent macrophages along differentiation as compared with RGD-coated surfaces, which were used as positive controls. Fg did not potentiate FBGC formation. In addition, actin cytoskeleton staining revealed the presence of punctuate F-actin with more elongated and interconnecting cells on Ch substrates. Antibody array screening and quantification of inflammation- and wound-healing-related factors indicated an overall reduction in Ch-based substrates versus RGD-coated surfaces. At late times, most inflammatory agents were down-regulated in the presence of Fg, in contrast to growth factor production, which was stimulated by Fg. Importantly, on Ch+Fg substrates, fully differentiated macrophages produced significant amounts of macrophage inflammatory protein-1delta (MIP-1δ), platelet-derived growth factor-BB, bone morphogenetic protein (BMP)-5, and BMP-7 compared with Ch alone. In addition, other important factors involved in bone homeostasis and wound healing, such as growth hormone, transforming growth factor-ß3, and insulin-like growth factor-binding proteins, as well as several angiogenic mediators, including endocrine gland-derived vascular endothelial factor, fibroblast growth factor-7, and placental growth factor, were significantly promoted by Fg. This work provides a new perspective on the inflammatory response in the context of bone repair/regeneration mediated by a pro-inflammatory protein (Fg) adsorbed onto a biomaterial (Ch) that does not otherwise exhibit osteogenic properties.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Bone and Bones/metabolism , Fibrinogen/pharmacology , Macrophages/metabolism , Monocytes/metabolism , Neovascularization, Physiologic/drug effects , Adsorption/drug effects , Cell Adhesion/drug effects , Cell Shape/drug effects , Cells, Cultured , Chitosan/pharmacology , Cytokines/metabolism , Giant Cells, Foreign-Body/drug effects , Giant Cells, Foreign-Body/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Macrophages/cytology , Macrophages/drug effects , Monocytes/cytology , Monocytes/drug effects
4.
Exp Mol Pathol ; 91(3): 673-81, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21798256

ABSTRACT

Foreign body-type multinucleated giant cells (FBGC), formed by macrophage fusion, are a prominent cell type on implanted biomaterials, although the roles they play at these and other sites of chronic inflammation are not understood. Why lymphocytes are present in this scenario and the effects of fusing macrophages/FBGC on subsequent lymphocyte responses are also unclear. To address the physiological significance of FBGC in this regard, we employed our in vitro system of interleukin (IL)-4-induced human monocyte-derived macrophage fusion/FBGC formation. Initially, we pursued the identities of lymphocyte co-stimulatory molecules on fusing macrophages/FBGC. In addition, we further compared the FBGC phenotype to that currently associated with osteoclasts and dendritic cells using recognized markers. Immunoblotting of cell lysates and immunochemistry of macrophages/FBGC in situ, revealed that IL-4-induced macrophages/FBGC strongly express HLA-DR, CD98, B7-2 (CD86), and B7-H1 (PD-L1), but not B7-1 (CD80) or B7-H2 (B7RP-1). Furthermore, molecules currently recognized to be expressed on osteoclasts (calcitonin receptor, tartrate-resistant acid phosphatase, RANK) or dendritic cells (CD1a, CD40, CD83, CD95/fas) are undetectable. In contrast, fusing macrophages/FBGC strongly express the macrophage markers αX integrin (CD11c), CD68, and dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), whereas CD14 is completely down-modulated with IL-4-induced macrophage fusion. These novel data demonstrate that IL-4-induction of macrophage multinucleation/FBGC formation features the acquisition of a CD14-negative phenotypic profile which is distinguishable from that of dendritic cells and osteoclasts, yet potentially exhibits multiple capacities for lymphocyte interactions with resultant lymphocyte down-modulation.


Subject(s)
Giant Cells, Foreign-Body , Acid Phosphatase/biosynthesis , Antigens, CD/biosynthesis , B7 Antigens/biosynthesis , Cell Adhesion Molecules/biosynthesis , Cell Fusion , Dendritic Cells/cytology , Dendritic Cells/metabolism , Giant Cells, Foreign-Body/cytology , Giant Cells, Foreign-Body/metabolism , Humans , Immunophenotyping , Interleukin-4 , Isoenzymes/biosynthesis , Lectins, C-Type/biosynthesis , Macrophage Activation , Macrophages/cytology , Monocytes/cytology , Osteoclasts/cytology , Osteoclasts/metabolism , Receptors, Calcitonin/biosynthesis , Receptors, Cell Surface/biosynthesis , Tartrate-Resistant Acid Phosphatase
5.
Adv Exp Med Biol ; 713: 97-111, 2011.
Article in English | MEDLINE | ID: mdl-21432016

ABSTRACT

Macrophages undergo fusion with other macrophages to form the hallmark multinucleated giant cells of chronic inflammation. However, neither the existence of distinct morphological types of giant cells, the signaling pathways that induce their formation, the molecular mechanism(s) of macrophage fusion, nor the significance of macrophage multinucleation at chronic inflammatory sites are well understood. Our efforts have been focused on these unknowns, particularly as they relate to the foreign body-type giant cells that form on implanted biomaterials and biomedical devices. We have pursued the discoveries of human macrophage fusion factors (interleukin-4, interleukin-13, α-tocopherol) with emphasis on foreign body giant cells, and identified adhesion receptors and signaling intermediates, as well as an adhesion protein substrate (vitronectin) that supports macrophage fusion. Studies on the molecular mechanism of macrophage fusion have revealed it to be a mannose receptor-mediated phagocytic process with participation of the endoplasmic reticulum. Further phenotypic and functional investigations will foster new perspectives on these remarkable multinucleated cells and their physiological significances in multiple inflammatory processes.


Subject(s)
Cell Fusion , Giant Cells, Foreign-Body/metabolism , Inflammation/immunology , Macrophages/physiology , Cell Adhesion/physiology , Cell Nucleus/metabolism , Giant Cells/cytology , Giant Cells/physiology , Giant Cells, Foreign-Body/cytology , Humans , Macrophages/cytology , Phenotype , Signal Transduction/physiology
6.
Semin Immunopathol ; 33(3): 221-33, 2011 May.
Article in English | MEDLINE | ID: mdl-21271251

ABSTRACT

The monocyte-derived macrophage is recognized as a critical determinant in biocompatibility, but its appearance in the chronic inflammatory phase is accompanied by the presence of lymphocytes, which have been much less studied in this regard. Here, we first present an overview of the physiologic continuum comprising host reactions to the surgical implantation of biomaterial. Secondly, we describe our collective research efforts, which indicate that lymphocytes are additional and key cellular determinants of biocompatible outcome. Thus, bioengineering advances will require that lymphocyte responses be regarded as integral components of innate inflammatory and immune/immunotoxic cell interactions at sites of biomaterial implantation.


Subject(s)
Biocompatible Materials , Lymphocytes/immunology , Macrophages/immunology , Prostheses and Implants , Animals , Cell Communication/immunology , Humans , Inflammation/immunology , Wound Healing/immunology
7.
J Biomed Mater Res A ; 94(3): 683-7, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20336743

ABSTRACT

The effect of polymorphonuclear leukocytes (PMNs) on the subsequent chronic phase macrophage-mediated foreign body reaction has not been previously investigated. Furthermore, while monocyte/macrophage-produced cytokines such as GM-CSF, G-CSF, or IL-1beta have been shown to increase PMN survival in vitro, few studies have examined the impact of directly cocultured monocytes/macrophages on PMN viability. To this end, we used our established in vitro system of interleukin (IL)-4-induced monocyte-derived macrophage fusion to examine the role of PMNs in the subsequent foreign body reaction. Monocytes were directly cultured with PMNs for 3 days before the addition of IL-4 to induce monocyte-derived macrophage fusion to facilitate foreign body giant cell (FBGC) formation by days 7 and 10 of culture. Optical microscopy was used to quantitatively determine adherent monocyte density, percent macrophage fusion, and FBGC density. A colorimetric MTT assay was used to assess PMN viability for direct cocultures of monocytes/macrophages and PMNs. Our results strongly suggest that the presence of PMNs inhibit IL-4-induced macrophage fusion and FBGC formation. Additionally, our findings demonstrate that cocultures containing PMNs and monocytes/macrophages increases PMN survival with respect to PMN-only cultures in vitro.


Subject(s)
Foreign-Body Reaction/immunology , Macrophages/immunology , Monocytes/immunology , Neutrophils/immunology , Cell Culture Techniques , Cells, Cultured , Coculture Techniques , Humans , Macrophages/cytology , Monocytes/cytology
8.
Exp Mol Pathol ; 84(1): 37-45, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18067888

ABSTRACT

Multinucleated giant cells are a classic cellular feature of chronic inflammation, although the mechanism of macrophage fusion leading to their formation is not well understood. Here, we investigate the participation of protein kinase C (PKC) in the interleukin (IL)-4-induced fusion of human monocyte-derived macrophages and foreign body giant cell (FBGC) formation in vitro. The PKC inhibitors H-7 and calphostin C attenuated macrophage fusion, whereas H-8, which is more selective for PKA and PKG, did not. Macrophage fusion was also prevented by the phospholipase C inhibitor, Et-18-OCH(3), the PKC isoform inhibitors GO6983 or rottlerin and by peptide inhibitors for PKC (20-28), PKCbeta, or PKCzeta but not by HBDDE or peptide inhibitors for PKCvarepsilon or PKA. In cultures of fusing macrophages/FBGC, we detected only PKCalpha, beta, delta, and zeta by immunoprecipitation and immunoblotting, and we also observed strong expression of these isoforms by immunocytochemistry. Our collective results suggest that the gamma, epsilon, eta, mu, theta, or iota PKC isoforms are not required in the mechanism of IL-4-induced macrophage fusion; whether PKCalpha is required is unclear. However, new evidence is provided that FBGC formation is supported by PKCbeta, PKCdelta, and PKCzeta in combined diacylglycerol-dependent (PKCbeta and PKCdelta) and -independent (PKCzeta) signaling pathways.


Subject(s)
Giant Cells, Foreign-Body/metabolism , Isoenzymes/metabolism , Protein Kinase C-delta/metabolism , Protein Kinase C/metabolism , Signal Transduction/physiology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/metabolism , Acetophenones/metabolism , Benzopyrans/metabolism , Carbazoles/metabolism , Cell Fusion , Cells, Cultured , Enzyme Inhibitors/metabolism , Giant Cells, Foreign-Body/cytology , Humans , Indoles , Isoenzymes/genetics , Isoquinolines/metabolism , Macrophages/cytology , Macrophages/metabolism , Maleimides , Monocytes/cytology , Monocytes/metabolism , Naphthalenes/metabolism , Protein Kinase C/genetics , Protein Kinase C beta , Protein Kinase C-delta/genetics
9.
J Biomed Mater Res A ; 84(1): 158-66, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17607751

ABSTRACT

Matrix metalloproteinases (MMPs) can degrade structural components within the extracellular matrix and at the cellular surface producing changes in cellular behavior (i.e., adhesion and migration) and subsequent pathological responses (i.e., the foreign body reaction and wound healing). We continue to study the foreign body reaction that occurs following biomaterial implantation by investigating secretory responses of biomaterial-adherent macrophages and foreign body giant cells (FBGCs) as directed by material surface chemistry and further this research by determining whether secreted MMPs play a role in macrophage adhesion and fusion. We have identified numerous MMPs and their tissue inhibitors (TIMPs) in in vitro cell-culture supernatants using antibody arrays and quantified select MMP/TIMPs with ELISAs. MMP-9 concentrations were significantly greater than both TIMP-1 and TIMP-2 on all materials. The ratios of MMP-9/TIMP-1 and MMP-9/TIMP-2 increased with time because of an increase in MMP-9 concentrations over time, while the TIMP concentrations remained constant. Total MMP-9 concentrations in the supernatants were comparable on all materials at each timepoint, while TIMP-1 and TIMP-2 concentrations tended to be greater on hydrophilic/anionic surfaces. Analysis of the MMP/TIMP quantities produced per cell revealed that the hydrophilic/neutral surfaces, which inhibited macrophage adhesion, activated the adherent macrophages/FBGCs to produce a greater quantity of MMP-9, TIMP-1, and TIMP-2 per cell. Pharmacological inhibition of MMP-1,-8,-13, and -18 reduced macrophage fusion without affecting adhesion, while inhibitors of MMP-2,-3,-9, and -12 did not affect adhesion or fusion. These findings demonstrate that material surface chemistry does modulate macrophage/FBGC-derived MMP/TIMP secretion and implicates MMP involvement in macrophage fusion.


Subject(s)
Biocompatible Materials/pharmacology , Foreign-Body Reaction/enzymology , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/metabolism , Tissue Inhibitor of Metalloproteinases/metabolism , Antibodies , Cells, Cultured , Enzyme Activation , Humans , Macrophages/drug effects , Macrophages/enzymology , Macrophages/metabolism , Matrix Metalloproteinases/immunology , Oligopeptides/metabolism , Protein Array Analysis , Substrate Specificity , Time Factors , Tissue Inhibitor of Metalloproteinases/immunology
10.
J Biomed Mater Res A ; 86(2): 535-43, 2008 Aug.
Article in English | MEDLINE | ID: mdl-17994558

ABSTRACT

An in vitro system of interleukin (IL)-4-induced foreign body giant cell (FBGC) formation was utilized to define the adhesion protein substrate(s) that promotes this aspect of the foreign body reaction on biomedical polymers. Human monocytes were cultured on cell culture polystyrene surfaces that had been pre-adsorbed with a synthetic arginine-glycine-aspartate peptide previously found to support optimal FBGC formation, or with various concentrations of potential physiological protein substrates, i.e. complement C3bi, collagen types I or IV, fibrinogen, plasma fibronectin, fibroblast fibronectin, laminin, thrombospondin, vitronectin, or von Willebrand factor. Cultures were evaluated on days 0 (1.5 h), 3, and 7 by May-Grünwald/Giemsa staining. Initial monocyte adhesion occurred on all adsorbed proteins. However, by day 7 of culture, only vitronectin was striking in its ability to support significant macrophage adhesion, development, and fusion leading to FBGC formation. Vitronectin supported high degrees of FBGC formation at an absorption concentration between 5 and 25 microg/mL. These findings suggest that adsorbed vitronectin is critical in the collective events that support and promote FBGC formation on biomedical polymers, and that the propensity for vitronectin adsorption may underlie the material surface chemistry dependency of FBGC formation.


Subject(s)
Cell Adhesion/drug effects , Giant Cells, Foreign-Body/cytology , Monocytes/cytology , Polystyrenes/pharmacology , Vitronectin/physiology , Biocompatible Materials , Cell Culture Techniques , Humans , Interleukin-4/pharmacology , Macrophages/cytology , Tissue Adhesions
11.
J Biomed Mater Res A ; 82(3): 568-74, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17311314

ABSTRACT

As beta1 and beta2 integrins were previously found to mediate adhesion during IL-4-induced foreign body giant cell (FBGC) formation, we pursued the identities of the alpha integrin partners of these adhesion receptors using our in vitro system of human monocyte-derived macrophage fusion. Immunoprecipitation with beta1 and immunoblotting reveal the presence of alpha5 and alphaV, as well as alpha2 and alpha3. alphaM and alphaX immunoprecipitate with beta2 but not with beta1. Immunocytochemistry coupled with confocal microscopy indicates that alpha5 and alphaX are poorly expressed on day 0. However, following the induction of fusion by IL-4 on day 3, they are each readily detectable in fusing macrophages/FBGC on day 7. In contrast, alphaM and alphaV are present throughout the culture period, with very strong alphaM expression on day 7. We also demonstrate expression and colocalization of alpha3, alpha5, or alphaV with beta1 on fusing macrophages/FBGC at this time point as well as strong colocalization of alphaM and alphaX with beta2 in FBGC and at fusion interfaces. Therefore, IL-4-induced FBGC are characterized by the expression of alphaMbeta2, alphaXbeta2, alpha5beta1, alphaVbeta1, alpha2beta1, and alpha3beta1, which indicates potential interactions with fragments of complement C3, fibrin(ogen), fibronectin, Factor X, and vitronectin, and possibly with certain collagens, laminin, and thrombospondin at sites of biomaterial implantation.


Subject(s)
CD18 Antigens/metabolism , Cell Fusion , Giant Cells, Foreign-Body/cytology , Integrin alpha Chains/metabolism , Integrin beta1/metabolism , Interleukin-4/pharmacology , Cell Adhesion , Cells, Cultured , Giant Cells, Foreign-Body/drug effects , Humans , Macrophages/cytology
12.
Exp Mol Pathol ; 79(2): 126-35, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16109404

ABSTRACT

Macrophage fusion leading to formation of multinucleated giant cells during chronic inflammation is poorly understood in mechanism and physiological significance. To address this, we developed a system of human macrophage fusion that utilizes IL-4, IL-13, or alpha-tocopherol to generate large foreign body-type giant cells (FBGC). Extending our previously demonstrated requirements for F-actin and mannose receptor (MR) activity, we find that macrophage fusion exhibits further features of a phagocytic process. Pharmacological inhibition of IL-4-induced FBGC formation indicates critical roles for vacuolar-type ATPase, microtubules, the endoplasmic reticulum (ER), and calcium-independent phospholipase A(2) (iPLA(2)), but not calcium-dependent PLA(2) (cPLA(2)), secretory PLA(2) (sPLA(2)), cyclooxygenase, or lipoxygenase. Immunocytochemistry confirms iPLA(2) expression and absence of cPLA(2) or sPLA(2) expression in macrophages/FBGC. As markers of ER-mediated phagocytosis, calnexin and calregulin are detectable on non-permeabilized fusing macrophages and also concentrated at fusion interfaces where they co-localize with actin in permeabilized macrophages/FBGC. Furthermore, ER markers co-localize with concanavalin A reactivity on non-permeabilized fusing macrophages, suggesting that the ER may present MR ligand during fusion events. These data demonstrate for the first time that the mechanism of macrophage fusion leading to formation of multinucleated giant cells exhibits multiple features of phagocytosis with potential participation of the ER.


Subject(s)
Cell Fusion/methods , Endoplasmic Reticulum/physiology , Giant Cells/physiology , Phagocytosis/physiology , Adenosine Triphosphatases/drug effects , Adenosine Triphosphatases/metabolism , Cells, Cultured , Concanavalin A/metabolism , Enzyme Inhibitors/pharmacology , Giant Cells/ultrastructure , Group VI Phospholipases A2 , Humans , Immunohistochemistry , Interleukin-13/metabolism , Interleukin-4/metabolism , Lectins, C-Type/metabolism , Lipoxygenase/drug effects , Lipoxygenase/metabolism , Macrophages/physiology , Macrophages/ultrastructure , Mannose Receptor , Mannose-Binding Lectins/metabolism , Microscopy, Confocal , Microtubules/drug effects , Microtubules/metabolism , Phagocytosis/drug effects , Phospholipases A/metabolism , Prostaglandin-Endoperoxide Synthases/drug effects , Prostaglandin-Endoperoxide Synthases/metabolism , Receptors, Cell Surface/metabolism
13.
Am J Pathol ; 163(3): 1147-56, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12937156

ABSTRACT

Multinucleated foreign body giant cells (FBGCs) form by monocyte-derived macrophage fusion on implanted biomedical devices and are believed to mediate oxidative damage to biomaterial surfaces. Our in vitro system of human macrophage culture and interleukin (IL)-4-induced FBGC formation was developed to study the macrophage fusion mechanism and the physiological significance of FBGCs on implanted biomaterials and at other sites of chronic inflammation. Here, we demonstrate that the antioxidant vitamin E (90% alpha-tocopherol) moderately induces macrophage fusion and increases IL-4-induced FBGC formation. Moreover, purified alpha-tocopherol, but not beta-, gamma-, or delta-tocopherol, most remarkably induces macrophage fusion, leading to cultures of confluent FBGCs below normal plasma concentrations. This is not observed with the similar antioxidants probucol or Trolox, suggesting that the alpha-tocopherol effects on FBGC formation are independent of its antioxidant activity. Consistent with the reported activation of diacylglycerol kinase by alpha-tocopherol, the diacylglycerol kinase inhibitor R59022 completely abrogates FBGC formation. R59022 inhibition of IL-4-induced FBGC formation is reversed by alpha-tocopherol, suggesting that FBGC formation involves diacylglycerol kinase activation. This study suggests a novel role for diacylglycerol kinase in the mechanism of macrophage fusion/FBGC formation at sites of chronic inflammation and reveals that the pleiotropic lipophilic compound, alpha-tocopherol, is a highly potent macrophage fusion factor.


Subject(s)
Diacylglycerol Kinase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Giant Cells, Foreign-Body/drug effects , Pyrimidinones/pharmacology , Thiazoles/pharmacology , alpha-Tocopherol/pharmacology , Antioxidants/pharmacology , Cell Adhesion/drug effects , Cell Fusion , Cells, Cultured , Drug Synergism , Humans , Interleukin-4/pharmacology , Macrophages/drug effects , Macrophages/physiology , Vitamin E/pharmacology
14.
Am J Pathol ; 160(2): 621-30, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11839583

ABSTRACT

An in vitro system of interleukin (IL)-4-induced human monocyte-derived macrophage fusion was used to investigate the cell/substrate adhesive mechanisms that support multinucleated foreign body giant cell (FBGC) formation. Monocytes were cultured for 3 days and IL-4 was added to induce macrophage fusion and FBGC formation by day 7. Functionally defined anti-integrin antibodies demonstrated that initial monocyte adhesion is mediated by beta2 integrins, whereas during the induction of macrophage fusion by IL-4, an additional dependence on beta1 integrins is acquired. The combination of anti-beta1 plus anti-beta2 was most effective, reducing macrophage/FBGC adhesion to 10% of controls. Consistent with integrin-mediated signaling, the tyrosine kinase inhibitor genistein and the phosphatidylinositol-3-kinase inhibitors wortmannin and LY294002 also attenuated macrophage/FBGC adhesion. Confocal microscopic analysis revealed that beta2 integrins are present on monocytes after initial adhesion and are strongly expressed on fusing macrophages, particularly in peripheral cell areas, and on FBGCs. In contrast, beta1 integrins are not detected on monocytes but begin to appear during macrophage development and are strongly expressed on fusing macrophages and FBGCs. For the first time, these results demonstrate the IL-4-induced acquisition of cooperation between beta1 and beta2 integrins in the cell/substrate adhesive interactions that are required for multinucleated FBGC formation.


Subject(s)
CD18 Antigens/metabolism , Cell Adhesion , Cell Fusion , Giant Cells, Foreign-Body/physiology , Integrin beta1/metabolism , Macrophages/physiology , Amino Acid Sequence , CD18 Antigens/pharmacology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Fibronectins/chemistry , Fibronectins/metabolism , Giant Cells, Foreign-Body/cytology , Giant Cells, Foreign-Body/drug effects , Humans , Immunohistochemistry , Integrin beta1/pharmacology , Interleukin-4/pharmacology , Macrophages/cytology , Macrophages/drug effects , Microscopy, Confocal , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Peptide Fragments/metabolism , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...