Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
Nutrients ; 11(10)2019 Sep 20.
Article in English | MEDLINE | ID: mdl-31547048

ABSTRACT

BACKGROUND: Recently, we demonstrated that Astragalus polysaccharide (PG2), the active ingredient in dried roots of astragalus membranaceus, ameliorates cancer symptom clusters and improves quality of life (QoL) in patients with metastatic disease by modulating inflammatory cascade against the background roles of inflammatory cells, including macrophages, dendritic cells (DCs), and cytotoxic T lymphocytes (CTLs) in tumor initiation, metastasis, and progression. Nevertheless, the role of PG2 in the modulation of anticancer immunogenicity and therapeutic response remains relatively underexplored and unclear. PURPOSE: The present study investigates how and to what extent PG2 modulates cellular and biochemical components of the inflammatory cascade and enhances anticancer immunity, as well as the therapeutic implication of these bio-events in patients with lung cancer. METHODS AND RESULTS: Herein, we demonstrated that PG2 significantly increased the M1/M2 macrophage polarization ratio in non-small cell carcinoma (NSCLC) H441 and H1299 cells. This PG2-induced preferential pharmacologic up-regulation of tumoral M1 population in vitro positively correlated with the downregulation of tumor-promoting IL-6 and IL-10 expression in NSCLC cell-conditioned medium, with concomitant marked inhibition of cell proliferation, clonogenicity, and tumorsphere formation. Our ex vivo results, using clinical sample from our NSCLC cohort, demonstrated that PG2 also promoted the functional maturation of DCs with consequent enhancement of T cell-mediated anticancer immune responses. Consistent with the in vitro and ex vivo results, our in vivo studies showed that treatment with PG2 elicited significant time-dependent depletion of the tumor-associated M2 population, synergistically enhanced the anti-M2-based anticancer effect of cisplatin, and inhibited xenograft tumor growth in the NSCLC mice models. Moreover, in the presence of PG2, cisplatin-associated dyscrasia and weight-loss was markedly suppressed. CONCLUSION: These results do indicate a therapeutically-relevant role for PG2 in modulating the M1/M2 macrophage pool, facilitating DC maturation and synergistically enhancing the anticancer effect of conventional chemotherapeutic agent, cisplatin, thus laying the foundation for further exploration of the curative relevance of PG2 as surrogate immunotherapy and/or clinical feasibility of its use for maintenance therapy in patients with lung cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Astragalus propinquus/chemistry , Lung Neoplasms/drug therapy , Macrophages/drug effects , Polysaccharides/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/pharmacology , Dendritic Cells/drug effects , Galectin 3/drug effects , Humans , Interleukin-10/metabolism , Interleukin-6/metabolism , Lung Neoplasms/immunology , Macrophage-1 Antigen/drug effects , Mice , Quality of Life , T-Lymphocytes/drug effects
2.
J Leukoc Biol ; 104(6): 1159-1171, 2018 12.
Article in English | MEDLINE | ID: mdl-30088666

ABSTRACT

Sepsis is a severe inflammatory condition associated with high mortality. Transmigration of neutrophils into tissues increases their lifespan to promote deleterious function. Junctional adhesion molecule-C (JAM-C) plays a pivotal role in neutrophil transmigration into tissues. We aim to study the role of JAM-C on the aging of neutrophils to cause sepsis-induced acute lung injury (ALI). Sepsis was induced in C57BL/6J mice by cecal ligation and puncture (CLP) and JAM-C expression in serum was assessed. Bone marrow-derived neutrophils (BMDN) were treated with recombinant mouse JAM-C (rmJAM-C) ex vivo and their viability was assessed. CLP-operated animals were administrated with either isotype IgG or anti-JAM-C Ab at a concentration of 3 mg/kg and after 20 h, aged neutrophils (CXCR4+ ) were assessed in blood and lungs and correlated with systemic injury and inflammatory markers. Soluble JAM-C level in serum was up-regulated during sepsis. Treatment with rmJAM-C inhibited BMDN apoptosis, thereby increasing their lifespan. CLP increased the frequencies of CXCR4+ neutrophils in blood and lungs, while treatment with anti-JAM-C Ab significantly reduced the frequencies of CXCR4+ aged neutrophils. Treatment with anti-JAM-C Ab significantly reduced systemic injury markers (alanine aminotransferase, aspartate aminotransferase, and lactate dehydrogenase) as well as systemic and lung inflammatory cytokines (IL-6 and IL-1ß) and chemokine (macrophage inflammatory protein-2). The blockade of JAM-C improved lung histology and reduced neutrophil contents in lungs of septic mice. Thus, reduction of the pro-inflammatory aged neutrophils by blockade of JAM-C has a novel therapeutic potential in sepsis-induced ALI.


Subject(s)
Acute Lung Injury/drug therapy , Cell Adhesion Molecules/antagonists & inhibitors , Molecular Targeted Therapy , Neutrophils/immunology , Sepsis/complications , Acute Lung Injury/etiology , Acute Lung Injury/immunology , Animals , Antibodies/therapeutic use , Antibody Specificity , Apoptosis , Cell Adhesion Molecules/blood , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/physiology , Cellular Senescence , Cytokines/blood , Immunoglobulin G/pharmacology , Immunoglobulins/blood , Immunoglobulins/immunology , Immunoglobulins/physiology , Lung/immunology , Lung/pathology , Macrophage-1 Antigen/drug effects , Macrophage-1 Antigen/physiology , Male , Mice , Mice, Inbred C57BL , Neutrophils/chemistry , Real-Time Polymerase Chain Reaction , Receptors, CXCR4/analysis , Recombinant Proteins/pharmacology , Transendothelial and Transepithelial Migration/drug effects
4.
Thromb Haemost ; 112(2): 379-89, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24652469

ABSTRACT

The co-stimulatory immune molecule CD40L figures prominently in a variety of inflammatory conditions including arterial disease. Recently, we made the surprising finding that CD40L mediates atherogenesis independently of its classic receptor CD40 via a novel interaction with the leukocyte integrin Mac-1. Here, we hypothesised that selective blockade of the CD40L-Mac-1 interaction may also retard restenosis. We induced neointima formation in C57/BL6 mice by ligation of the left carotid artery. Mice were randomised to daily intraperitoneal injections of either cM7, a small peptide selectively inhibiting the CD40L-Mac-1 interaction, scM7, a scrambled control peptide, or saline for 28 days. Interestingly, cM7-treated mice developed neointima of similar size compared with mice receiving the control peptide or saline as assessed by computer-assisted analysis of histological cross sections. These data demonstrate that the CD40L-Mac-1 interaction is not required for the development of restenosis. In contrast, CD40-deficient mice subjected to carotid ligation in parallel, developed significantly reduced neointimal lesions compared with respective wild-type controls (2872 ± 843 µm² vs 35469 ± 11870 µm²). Flow cytometry in CD40-deficient mice revealed reduced formation of platelet-granulocyte and platelet-inflammatory monocyte- aggregates. In vitro, supernatants of CD40-deficient platelet-leukocyte aggregates attenuated proliferation and increased apoptosis of smooth muscle cells. Unlike in the setting of atherosclerosis, CD40L mediates neointima formation via its classic receptor CD40 rather than via its recently described novel interaction with Mac-1. Therefore, selective targeting of CD40L-Mac-1 binding does not appear to be a favorable strategy to fight restenosis.


Subject(s)
CD40 Antigens/metabolism , CD40 Ligand/antagonists & inhibitors , Carotid Arteries/drug effects , Carotid Stenosis/prevention & control , Macrophage-1 Antigen/drug effects , Neointima , Oligopeptides/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis , CD40 Antigens/immunology , CD40 Ligand/genetics , CD40 Ligand/immunology , CD40 Ligand/metabolism , Carotid Arteries/immunology , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Stenosis/immunology , Carotid Stenosis/metabolism , Carotid Stenosis/pathology , Cells, Cultured , Disease Models, Animal , Leukocyte Rolling/drug effects , Macrophage-1 Antigen/immunology , Macrophage-1 Antigen/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Platelet Activation/drug effects , Recurrence
5.
Am J Physiol Regul Integr Comp Physiol ; 306(7): R483-9, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24500432

ABSTRACT

The inflammatory response to muscle-damaging exercise requires monocyte mobilization and adhesion. Complement receptor type 3 (CR3) and macrophage inflammatory protein (MIP)-1ß enables monocyte recruitment, adhesion, and subsequent infiltration into damaged muscle tissue. The purpose of this study was to examine the effects of cold water immersion (CWI) and/or ß-hydroxy-ß-methylbutyrate free acid (HMB-FA) on CR3 expression and MIP-1ß concentration after four sets of up to 10 repetitions of squat, dead lift, and split squat exercises at 70-80% 1-repetition maximum. Thirty-nine resistance-trained men (22.2 ± 2.5 yr) were randomly divided into four groups: 1) placebo (PL), 2) HMB-FA, 3) HMB-FA-CWI, and 4) PL-CWI. The HMB-FA groups ingested 3 g/day, and CWI groups were submersed into 10-12°C water for 10 min after exercise. Blood was sampled at baseline (PRE), immediately post- (IP), 30 min post- (30P), 24 h post- (24P), and 48 h post (48P)-exercise. Circulating MIP-1ß was assayed and CR3 expression on CD14+ monocytes was measured by flow cytometry. Without treatment, CR3 expression significantly elevated at 30P compared with other time points (P = 0.030-0.047). HMB-FA significantly elevated the percentage of monocytes expressing CR3 between IP and 24P (P = 0.046) and between IP and 48P (P = 0.046). No time effect was observed for MIP-1ß concentration. The recovery modalities showed to attenuate the rise in CR3 following exercise. Additionally, supplementation with HMB-FA significantly elevated the percentage of monocytes expressing CR3 during recovery. Although the time course that inflammatory responses are most beneficial remains to be determined, recovery modalities may alter immune cell mobilization and adhesion mechanisms during tissue recovery.


Subject(s)
Chemokine CCL4/blood , Cold Temperature , Immersion , Macrophage-1 Antigen/drug effects , Monocytes/drug effects , Muscle Contraction , Muscle, Skeletal/drug effects , Resistance Training , Valerates/pharmacology , Water , Adult , Double-Blind Method , Florida , Humans , Macrophage-1 Antigen/blood , Male , Monocytes/immunology , Monocytes/metabolism , Muscle, Skeletal/immunology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Recovery of Function , Time Factors , Young Adult
6.
J Periodontol ; 85(8): 1096-106, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24354650

ABSTRACT

BACKGROUND: Chewing of areca quid increases the prevalence of periodontal diseases. Areca nut extract (ANE) inhibits the phagocytic activity of human neutrophils. This in vitro study investigates the effects of ANE on complement- and antibody-opsonized phagocytosis by neutrophils. Expression of complement receptors, Fc receptors, and F-actin in ANE-treated neutrophils is also analyzed. METHODS: The viability of ANE-treated neutrophils was determined using the propidium iodide staining method. The possible effects of ANE on the expression of complement receptors and Fc receptors were examined using an immunofluorescence staining method followed by flow cytometry and confocal laser scanning microscopy. The phagocytic activity of neutrophils against complement or immunoglobulin (Ig)G-opsonized fluorescent beads was analyzed using flow cytometry. Expression of F-actin was determined using confocal laser scanning microscopy. RESULTS: ANE significantly inhibited the production of complement receptors (CR1, CR3, and CR4) and Fc receptors (FcγRII and FcγRIII) in a concentration-dependent manner. Treatment of neutrophils with ANE significantly impaired their ability to phagocytose fluorescent beads. ANE also inhibited phagocytosis of fluorescent beads that were opsonized by complement or IgG. Moreover, expression of F-actin was inhibited after ANE treatment. CONCLUSIONS: ANE inhibits the complement- and IgG-mediated neutrophil phagocytosis that may result from reduction of the expression of complement receptors, Fc receptors, and F-actin formation after ANE treatment. The findings suggest that areca nut chewing may jeopardize the defensive functions of neutrophils and affect periodontal health.


Subject(s)
Areca , Neutrophils/drug effects , Nuts , Plant Extracts/pharmacology , Receptors, Complement/drug effects , Receptors, Fc/drug effects , Actins/drug effects , Adult , Cell Survival/drug effects , Cells, Cultured , Coloring Agents , Complement C1/drug effects , Female , Flow Cytometry , Fluorescent Antibody Technique, Direct , Humans , Integrin alphaXbeta2/drug effects , Macrophage-1 Antigen/drug effects , Male , Microscopy, Confocal , Microspheres , Neutrophils/immunology , Phagocytosis/drug effects , Propidium , Receptors, IgG/drug effects , Young Adult
7.
Blood ; 122(20): 3473-81, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-23878142

ABSTRACT

Interleukin-1ß (IL-1ß) is a proinflammatory cytokine and a therapeutic target in several chronic autoimmune states. Monocytes and macrophages are the major sources of IL-1ß. IL-1ß production by these cells requires Toll-like receptor (TLR) and adenosine triphosphate (ATP)-mediated P2X purinoceptor 7 (P2X7) signals, which together activate the inflammasome. However, how TLR signals and ATP availability are regulated during monocyte activation is unclear and the involvement of another danger signal system has been proposed. Here, we demonstrate that both lipopolysaccharide (LPS) and the anaphylatoxin C3a are needed for IL-1ß production in human macrophages and dendritic cells, while in monocytes, C3a enhanced the secretion of LPS-induced IL-1ß. C3a and LPS-stimulated monocytes increased T helper 17 (Th17) cell induction in vitro, and human rejecting, but not nonrejecting, kidney transplant biopsies were characterized by local generation of C3a and monocyte and Th17 cell infiltration. Mechanistically, C3a drives IL-1ß production in monocytes by controlling the release of intracellular ATP into the extracellular space via regulation of as-yet unidentified ATP-releasing channels in an extracellular signal-regulated kinase 1/2-dependent fashion. These data define a novel function for complement in inflammasome activation in monocytes and suggest that C3aR-mediated signaling is a vital component of the IL-1ß-Th17 axis.


Subject(s)
Adenosine Triphosphate/metabolism , Carrier Proteins/physiology , Complement C3/physiology , Inflammasomes/physiology , Interleukin-1beta/metabolism , Monocytes/metabolism , Cells, Cultured , Complement C3/agonists , Cytokines/biosynthesis , Cytokines/genetics , Dendritic Cells/metabolism , Enzyme Activation , Graft Rejection/immunology , Graft Rejection/pathology , Humans , Kidney Transplantation , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System , Macrophage-1 Antigen/drug effects , Macrophage-1 Antigen/physiology , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Purinergic P2X7/physiology , Recombinant Proteins/pharmacology , Th17 Cells/metabolism , Toll-Like Receptors/physiology
8.
Cell Adh Migr ; 5(1): 29-33, 2011.
Article in English | MEDLINE | ID: mdl-20935452

ABSTRACT

Magnetic nanoparticles surface-covered with meso-2,3-dimercaptosuccinic acid (MNPs-DMSA) constitute a promising approach for tissue- and cell-targeted delivery of therapeutic drugs in the lung. However, they can also induce a transient transendothelial migration of leukocytes in the organ as a side effect after endovenous administration of MNPs-DMSA. We demonstrated that monocytes/macrophages constitute the main subpopulation of leukocytes involved in this process. Our recent research found that MNPs-DMSA up-regulated the mRNA expression of E-, L- and P-selectin and macrophage-1 antigen, and increased concentration of tumor necrosis factor-α in lung, in a time dependent manner. The critical relevance of the ß2 integrin-dependent pathway in leukocyte transmigration elicited by MNPs-DMSA was demonstrated by use of knockout mice. Our work characterizes mechanisms of the pro-inflammatory effects of MNPs-DMSA in the lung, and identifies ß2 integrin-targeted interventions as promising strategies to reduce pulmonary side effects of MNPs-DMSA during biomedical applications. In addition, MNPs-DMSA could be used as modulators of lung immune response.


Subject(s)
Leukocytes/drug effects , Lung/drug effects , Magnetite Nanoparticles/administration & dosage , Succimer/administration & dosage , Transendothelial and Transepithelial Migration/drug effects , Animals , CD18 Antigens/metabolism , Leukocytes/metabolism , Macrophage-1 Antigen/drug effects , Macrophage-1 Antigen/metabolism , Macrophages/drug effects , Mice , P-Selectin/drug effects , P-Selectin/metabolism , Succimer/chemistry , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
9.
J Immunol ; 183(10): 6460-8, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19864611

ABSTRACT

Intracellular signals associated with or triggered by integrin ligation can control cell survival, differentiation, proliferation, and migration. Despite accumulating evidence that conformational changes regulate integrin affinity to its ligands, how integrin structure regulates signal transmission from the outside to the inside of the cell remains elusive. Using fluorescence resonance energy transfer, we addressed whether conformational changes in integrin Mac-1 are sufficient to transmit outside-in signals in human neutrophils. Mac-1 conformational activation induced by ligand occupancy or activating Ab binding, but not integrin clustering, triggered similar patterns of intracellular protein tyrosine phosphorylation, including Akt phosphorylation, and inhibited spontaneous neutrophil apoptosis, indicating that global conformational changes are critical for Mac-1-dependent outside-in signal transduction. In neutrophils and myeloid K562 cells, ligand ICAM-1 or activating Ab binding promoted switchblade-like extension of the Mac-1 extracellular domain and separation of the alpha(M) and beta(2) subunit cytoplasmic tails, two structural hallmarks of integrin activation. These data suggest the primacy of global conformational changes in the generation of Mac-1 outside-in signals.


Subject(s)
Leukocytes, Mononuclear/immunology , Macrophage-1 Antigen/immunology , Neutrophils/immunology , Signal Transduction/immunology , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Apoptosis/immunology , Carcinogens/pharmacology , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Line, Tumor , Humans , Immunologic Factors/pharmacology , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/metabolism , Interleukin-8/pharmacology , Leukemia/immunology , Leukemia/metabolism , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Macrophage-1 Antigen/drug effects , Macrophage-1 Antigen/metabolism , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Neutrophils/metabolism , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/physiology , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology
10.
J Neuroimmunol ; 214(1-2): 78-82, 2009 Sep 29.
Article in English | MEDLINE | ID: mdl-19656578

ABSTRACT

LPS activates microglia, which are normally maintained in a quiescent state by CD200-CD200 receptor (CD200R) interaction. MAC-1 (a microglia marker) mRNA expression was increased in mice brains up to 1 year post LPS administration (i.p.). Minocycline treatment did not prevent LPS (5 mg/kg)-induced increase in MAC-1 mRNA but reduced that induced by 0.1 mg/kg LPS. CD200R mRNA decreased starting at 4 h, whereas CD200 mRNA increased at 4 h and decreased at 1 year post LPS inoculation. Thus, LPS-induced changes in CD200-CD200R equilibrium might keep microglia chronically activated. Minocycline does not effectively inhibit microglia activation induced by high-dose LPS.


Subject(s)
Antigens, CD/metabolism , Brain/immunology , Macrophage-1 Antigen/metabolism , Membrane Glycoproteins/metabolism , Microglia/immunology , Animals , Anti-Bacterial Agents/pharmacology , Antigens, CD/drug effects , Brain/cytology , Brain/drug effects , Gene Expression/drug effects , Lipopolysaccharides , Macrophage-1 Antigen/drug effects , Membrane Glycoproteins/drug effects , Mice , Mice, Inbred C57BL , Microglia/drug effects , Minocycline/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
11.
Bioorg Med Chem ; 17(2): 558-68, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19097798

ABSTRACT

Three series of non-steroidal anti-inflammatory drugs (NSAIDs) inhibiting the cyclooxygenase/5-lipoxygenase (COX/5-LOX) pathways as such as formation of hydroxyl radicals and adhesion were prepared: 4,5-diaryl isothiazoles, 4,5-diaryl 3H-1,2-dithiole-3-thiones and 4,5-diaryl 3H-1,2-dithiole-3-ones. The aim of the present study was to develop substances which can intervene into the inflammatory processes via different mechanisms of action as multiple target non-steroidal anti-inflammatory drugs (MTNSAIDs) with increased anti-inflammatory potential. The current lead 11a was evaluated in COX-1/2, 5-LOX and (*)OH scavenging in vitro assays and in a static adhesion assay where it proved to inhibit adhesion. Moreover, 11a treatment attenuated expression of macrophage adhesion molecule-1 (Mac-1) on extravasated polymorphonuclear leukocytes (PMNs) which indicates that the activation was reduced. The assays used are predictive for the in vivo efficacy of test compounds as shown for 11a in a peritonitis model of acute inflammation in mice. Thus, the novel 5-LOX/COX and (*)OH inhibitor 11a possesses anti-inflammatory activity that, in addition to COX/5-LOX inhibition, implicates effects on leukocyte-endothelial interactions.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cyclooxygenase Inhibitors/chemistry , Lipoxygenase Inhibitors , Sulfhydryl Compounds/chemistry , Thiazoles/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Adhesion/drug effects , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase Inhibitors/pharmacology , Free Radical Scavengers , Hydroxyl Radical , Macrophage-1 Antigen/analysis , Macrophage-1 Antigen/drug effects , Mice , Neutrophils/drug effects , Peritonitis/drug therapy , Sulfhydryl Compounds/pharmacology , Thiazoles/pharmacology
12.
J Pharm Pharmacol ; 60(11): 1473-9, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18957168

ABSTRACT

This study was conducted to investigate putative antagonism of integrin receptors alphaMbeta2 and alphaLbeta2 by a novel coumarin derivative (BOL-303225-A), its efficacy in-vivo after retinal ischaemia-reperfusion injury, and its bioavailability in rat plasma. A cellular adhesion assay in Jurkat and U937 cells, and a flow cytometry assay with an antibody against the beta2 subunit were conducted. BOL-303225-A bioavailability in rat plasma and the retinal levels of myeloperoxidase (MPO) after ischaemia-reperfusion injury were evaluated after oral administration (10 mg kg(-1)). In-vitro cell viability assays revealed no cytotoxicity for BOL-303225-A over a wide dose range, and IC50 values of 32.3 +/- 1.5 muM and 84.95 +/- 2.3 muM were found for Jurkat and U937 cells, respectively. The drug showed specific binding to the alphaMbeta2 and alphaLbeta2 integrin receptors expressed by U937 and Jurkat cells, respectively, producing a fluorescence shift towards lower values in a concentration-dependent manner. The pharmacokinetic profile of BOL-303225-A exhibited rapid absorption following oral administration in the rat. A significant reduction of retinal MPO levels was observed in drug-treated rats. This study demonstrated that BOL-303225-A acts as an antagonist of the integrin alphaLbeta2 and alphaMbeta2 receptors, suggesting that this drug could be used for ocular diseases such as diabetic retinopathy.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Coumarins/pharmacology , Lymphocyte Function-Associated Antigen-1/drug effects , Macrophage-1 Antigen/drug effects , Administration, Oral , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Biological Availability , Cell Adhesion/drug effects , Cell Survival/drug effects , Coumarins/administration & dosage , Coumarins/pharmacokinetics , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Jurkat Cells , Lymphocyte Function-Associated Antigen-1/metabolism , Macrophage-1 Antigen/metabolism , Male , Peroxidase/drug effects , Peroxidase/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Retinal Diseases/drug therapy , Retinal Diseases/pathology , U937 Cells
13.
FASEB J ; 22(12): 4109-16, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18708587

ABSTRACT

The molecular and structural basis of anesthetic interactions with conformations and functionalities of cell surface receptors remains to be elucidated. We have demonstrated that the widely used volatile anesthetic isoflurane blocks the activation-dependent conformational conversion of integrin lymphocyte function associated antigen-1 (LFA-1), the major leukocyte cell adhesion molecule, to a high-affinity configuration. Perturbation of LFA-1 activation by isoflurane at clinically relevant concentrations leads to the inhibition of T-cell interactions with target cells as well as ligand-triggered intracellular signaling. Nuclear magnetic resonance spectroscopy reveals that isoflurane binds within a cavity in the LFA-1 ligand-binding domain, which is a previously identified drug-binding site for allosteric small-molecule antagonists that stabilize LFA-1 in a low-affinity conformation. These results provide a potential mechanism for the immunomodulatory properties of isoflurane.


Subject(s)
Allosteric Site/drug effects , Anesthetics, Inhalation/chemistry , Anesthetics, Inhalation/pharmacology , Isoflurane/chemistry , Isoflurane/pharmacology , Lymphocyte Function-Associated Antigen-1/chemistry , Humans , Intercellular Adhesion Molecule-1/metabolism , Jurkat Cells , K562 Cells , Lymphocyte Function-Associated Antigen-1/metabolism , Macrophage-1 Antigen/drug effects , Macrophage-1 Antigen/metabolism , Models, Biological , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation/drug effects , Up-Regulation/drug effects
14.
Immunopharmacol Immunotoxicol ; 29(3-4): 549-62, 2007.
Article in English | MEDLINE | ID: mdl-18075864

ABSTRACT

C3 binding glycoprotein (C3bgp) is immunomodulating molecule isolated from the plant Cuscuta europea. When neutrophils were incubated with C3bgp the subsequent binding of anti-CD11b mAb became significantly higher. C3bgp induced moderate TNF-alpha production in human PBMC and primary monocytes. This production was significantly inhibited by the specific inhibitors of JNK and p38 MAPKs. The inhibition of JNK reduced PBMC viability. We concluded that: (i) C3bgp utilized CD11b polypeptide chain of CR3 and mediated a part of its immunomodulatory properties by activation of JNK and p38 and (ii) PBMC viability at in vitro conditions depends of JNK signal transduction pathway activation.


Subject(s)
Carrier Proteins/pharmacology , Glycoproteins/pharmacology , Immunologic Factors , Macrophage-1 Antigen/drug effects , Mitogen-Activated Protein Kinases/physiology , Signal Transduction/drug effects , CD11b Antigen/metabolism , Cell Survival/drug effects , Enzyme-Linked Immunosorbent Assay , Humans , Lipopolysaccharides/pharmacology , MAP Kinase Kinase 4/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Phagocytes/drug effects , Tumor Necrosis Factor-alpha/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Scand J Immunol ; 65(4): 344-52, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17386025

ABSTRACT

Experimental toxocariasis was used as a model of eosinophil migration. Mice inoculated with 200 Toxocara canis eggs were treated with the leukotriene inhibitor MK886 (1 mg/kg/day). Eosinophils were counted in peripheral blood (PB), peritoneal cavity (PC) and bronchoalveolar lavage fluid (BALF) samples on post-infection days 3, 6, 12, 18, 24 and 36. Eosinophil expression of Mac-1 and VLA-4 was analysed in PB and PC samples. We found that T. canis infection induced systemic eosinophilia from post-infection day 3, peaking on days 6, 12 and 24 in PB, PC and BALF samples respectively. Eosinophilia was more pronounced in PB and PC samples than in BALF samples, and MK886 downregulated eosinophilia to varying degrees in the different sample types. In PB and PC samples, T. canis infection caused early upregulation of Mac-1 with late changes in the VLA-4 profile, whereas MK886 had opposite effects. The distinct time-dependent eosinophilia peaks and differential involvement of leukotrienes in integrin expression demonstrate that, despite the systemic eosinophilia triggered by T. canis infection, inflammatory responses vary by compartment.


Subject(s)
Eosinophilia/drug therapy , Eosinophils/drug effects , Indoles/therapeutic use , Lipoxygenase Inhibitors/therapeutic use , Toxocariasis/drug therapy , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Movement/drug effects , Cell Movement/immunology , Eosinophilia/immunology , Eosinophils/immunology , Female , Flow Cytometry , Integrin alpha4beta1/biosynthesis , Integrin alpha4beta1/drug effects , Macrophage-1 Antigen/biosynthesis , Macrophage-1 Antigen/drug effects , Mice , Mice, Inbred BALB C , Phenotype , Toxocariasis/immunology
16.
Glia ; 53(5): 538-50, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16374778

ABSTRACT

Complement-receptor-3 (CR3/MAC-1), scavenger-receptor-AI/II (SRAI/II), and Fcgamma-receptor (FcgammaR) can mediate myelin phagocytosis in macrophages and microglia. Paradoxically, after injury to CNS axons these receptors are expressed but myelin is not phagocytosed, suggesting that phagocytosis is subject to regulation between efficient and inefficient states. In the present work, we focus on CR3/MAC-1 and SRAI/II-mediated myelin phagocytosis. Phagocytosis by CR3/MAC-1 and SRAI/II was inhibited by cPKC inhibitor Go-6976, general-PKC inhibitors Ro-318220 and calphostin-C, and BAPTA/AM, which chelates intracellular Ca2+ required for cPKC activation. Signaling/activation by cPKC are thus suggested. PMA, which mimics diacylglycerol (DAG) as an activator of cPKC, novel-PKC (nPKC), and non-PKC DAG-driven molecule(s), produced a dose-dependent dual effect on phagocytosis by CR3/MAC-1 and SRAI/II, i.e., augmentation at low concentrations and inhibition at high concentrations. Inhibition of phagocytosis by CR3/MAC-1 was enhanced by combining inhibiting concentrations of PMA with PKC inhibitors Go-6976 or Ro-318220, suggesting inhibition by PMA/DAG-driven non-PKC molecule(s). In contrast, inhibition of phagocytosis by SRAI/II was enhanced by combining inhibiting concentrations of PMA with cPKC inhibitor Go-6976 but not with general-PKC inhibitor Ro-318220, suggesting inhibition by nPKC. Phagocytosis by CR3/MAC-1 and SRAI/II was further inhibited by PI3K inhibitors wortmannin and LY-294002 and PLCgamma inhibitor U-73122. Altogether, our observations suggest that CR3/MAC-1 and SRAI/II-mediated myelin phagocytosis share activation by PI3K, PLCgamma and cPKC. The two differ, however, in that non-PKC DAG-driven molecule(s) inhibit CR3/MAC-1-mediated phagocytosis, whereas nPKC inhibit SRAI/II-mediated phagocytosis. Each of these signaling steps may be targeted for regulating CR3/MAC-1 and/or SRAI/II-mediated phagocytosis between efficient and inefficient states.


Subject(s)
Macrophage-1 Antigen/drug effects , Myelin Sheath/immunology , Phagocytosis/physiology , Phosphatidylinositol 3-Kinases/pharmacology , Phospholipase C gamma/pharmacology , Protein Kinase C/pharmacology , Scavenger Receptors, Class A/antagonists & inhibitors , Animals , Chelating Agents/pharmacology , Dose-Response Relationship, Drug , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Activation/physiology , Enzyme-Linked Immunosorbent Assay , Immunoblotting , Immunoprecipitation , Isoenzymes/pharmacology , Macrophages, Peritoneal/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Microglia/physiology , Receptors, Drug , Signal Transduction/drug effects , Signal Transduction/physiology , Tetradecanoylphorbol Acetate/pharmacology , Thioglycolates/pharmacology , Tyrosine/metabolism
17.
Rev Cardiovasc Med ; 7 Suppl 4: S3-11, 2006.
Article in English | MEDLINE | ID: mdl-17224889

ABSTRACT

The platelet glycoprotein (GP) IIb/IIIa inhibitors differ markedly in their pharmacokinetics, pharmacodynamics, and differential receptor affinities. Abciximab and the small-molecule GP IIb/IIIa inhibitors (eptifibatide, tirofiban) have separate, distinct binding sites on the GP IIb/IIIa receptor complex. The affinity of abciximab for the platelet GP IIb/IIIa integrin receptor, together with non-platelet-receptor mediated effects achieved through its affinity for the aVb3 and CD11b/18 receptors, most likely contribute to the clinical benefit associated with the use of abciximab as adjunctive pharmacotherapy during primary percutaneous coronary intervention for the treatment of ST-elevation acute myocardial infarction.


Subject(s)
Angioplasty, Balloon, Coronary/adverse effects , Anti-Inflammatory Agents/therapeutic use , Blood Platelets/drug effects , Coronary Circulation/drug effects , Myocardial Infarction/therapy , Platelet Aggregation Inhibitors/therapeutic use , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Vasculitis/prevention & control , Abciximab , Anti-Inflammatory Agents/pharmacology , Antibodies, Monoclonal/therapeutic use , Blood Platelets/metabolism , Humans , Immunoglobulin Fab Fragments/therapeutic use , Integrin alphaVbeta3/antagonists & inhibitors , Macrophage-1 Antigen/drug effects , Microcirculation/drug effects , Myocardial Infarction/blood , Myocardial Infarction/drug therapy , Myocardial Infarction/physiopathology , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Vasculitis/etiology
19.
J Lipid Res ; 46(3): 475-83, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15576843

ABSTRACT

Two subfamilies of the polar glycopeptidolipids (GPLs) located on the surface of Mycobacterium smegmatis, along with unknown phospholipids, were recently shown to participate in the nonopsonic phagocytosis of mycobacteria by human macrophages (Villeneuve, C., G. Etienne, V. Abadie, H. Montrozier, C. Bordier, F. Laval, M. Daffe, I. Maridonneau-Parini, and C. Astarie-Dequeker. 2003. Surface-exposed glycopeptidolipids of Mycobacterium smegmatis specifically inhibit the phagocytosis of mycobacteria by human macrophages. Identification of a novel family of glycopeptidolipids. J. Biol. Chem. 278: 51291-51300). As demonstrated herein, a phospholipid mixture that derived from the methanol-insoluble fraction inhibited the phagocytosis of M. smegmatis. Inhibition was essentially attributable to phosphatidylinositol mannosides (PIMs), namely PIM2 and PIM6, because the purified phosphatidylethanolamine, phosphatidylglycerol, and phosphatidylinositol were inactive. This was further confirmed using purified PIM2 and PIM6 from M. bovis BCG that decreased by half the internalization of M. smegmatis. Both compounds also inhibited the uptake of M. tuberculosis and M. avium but had no effect on the internalization of zymosan used as a control particle of the phagocytic process. When coated on latex beads, PIM2 and polar GPL (GPL III) favored the particle entry through complement receptor 3. GPL III, but not PIM2, also directed particle entry through the mannose receptor. Therefore, surface-exposed mycobacterial PIM and polar GPL participate in the receptor-dependent internalization of mycobacteria in human macrophages.


Subject(s)
Glycolipids/pharmacology , Macrophage-1 Antigen/metabolism , Macrophages/drug effects , Macrophages/microbiology , Mycobacterium smegmatis/chemistry , Phagocytosis/drug effects , Carbohydrate Sequence , Cells, Cultured , Glycolipids/chemistry , Glycolipids/isolation & purification , Humans , Macrophage-1 Antigen/drug effects , Macrophages/immunology , Mass Spectrometry , Molecular Sequence Data , Molecular Structure , Mycobacterium bovis/chemistry , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/physiology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/physiology , Phosphatidylinositols/chemistry , Phosphatidylinositols/pharmacology
20.
J Am Coll Cardiol ; 44(7): 1408-14, 2004 Oct 06.
Article in English | MEDLINE | ID: mdl-15464320

ABSTRACT

OBJECTIVES: The aim of this study was to confirm clinically a hypothesis that cilostazol inhibits leukocyte Mac-1, leading to prevention of post-stent restenosis. BACKGROUND: The platelet phosphodiesterase III inhibitor called cilostazol also inhibits alpha-granule release of P-selectin in platelets. The P-selectin-mediated platelet-leukocyte interaction promotes activation and upregulation of leukocyte Mac-1 after coronary stenting, which plays a key role on the mechanism of restenosis. Thus, cilostazol's potential inhibition of this process may lead to prevention of restenosis. METHODS: Using flow cytometric analysis of whole blood obtained from the coronary sinus, the expression of platelet membrane glycoproteins and neutrophil adhesion molecules was observed in 70 consecutive patients undergoing coronary stenting. The patients were randomly assigned to either a cilostazol or ticlopidine group before stent placement. RESULTS: The restenosis rate was lower (15% vs. 31%, p < 0.05) in the cilostazol group (n = 34) than in the ticlopidine group (n = 32). A stent-induced increase in platelet P-selectin (CD62P) expression and an increase in neutrophil Mac-1 (CD11b) expression were suppressed in the cilostazol group compared with the ticlopidine group. Angiographic late lumen loss was correlated with the relative changes in platelet P-selectin and neutrophil Mac-1 at 48 h after coronary stenting. CONCLUSIONS: Cilostazol may have effects on suppression of P-selectin-mediated platelet activation, platelet-leukocyte interaction, and subsequent Mac-1-mediated leukocyte activation, which might lead to a reduced restenosis rate after coronary stent implantation.


Subject(s)
Coronary Artery Disease/therapy , Coronary Restenosis/prevention & control , Leukocytes/drug effects , Macrophage-1 Antigen/drug effects , Platelet Activation/drug effects , Stents , Tetrazoles/therapeutic use , Ticlopidine/therapeutic use , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Aged , Cilostazol , Coronary Angiography , Coronary Artery Disease/blood , Coronary Artery Disease/drug therapy , Coronary Restenosis/blood , Cyclic Nucleotide Phosphodiesterases, Type 3 , Female , Flow Cytometry , Humans , Leukocytes/metabolism , Male , Middle Aged , Neutrophils/drug effects , P-Selectin/blood , P-Selectin/drug effects , Platelet Aggregation Inhibitors/therapeutic use , Research Design , Stents/adverse effects , Tetrazoles/pharmacology , Ticlopidine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...