Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Ultraschall Med ; 30(2): 175-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18496776

ABSTRACT

PURPOSE: Ultrasound of the neck detects a large number of non-palpable thyroid nodules in the population, but it offers poor diagnostic accuracy (the presence of microcalcifications is the only statistically significant criterion indicative of malignancy). The aim of this study is to evaluate elastography, a technique which allows differentiation between pathological and normal tissue by determining its hardness and which could also prove useful in the characterisation of thyroid nodules. MATERIALS AND METHODS: In this prospective study, 51 thyroid nodules in 40 consecutive patients were examined (25 women, 15 men, mean age +/- SD, 54 +/- 13.4). Elastosonography was performed by real-time, free-hand technique, using Logos HiVision equipment with a 10 MHz transducer and lesions were classified and scored in 4 classes of hardness. All patients were also examined by grey scale high frequency ultrasound and colour Doppler. Final diagnoses were obtained from cytological and/or histological evaluation. RESULTS: Final diagnoses revealed 11 malignant and 40 benign nodules. Only in two cases ultrasound demonstrated signs useful for a differential diagnosis (intrinsic microcalcifications). Correct differentiation of malignant from benign nodules was obtained by elastosonography in 43 / 51 cases with 5 false positives (FP) and 3 false negatives (FN). Specificity, sensitivity and accuracy were 87.5 %, 81.8 % and 86.2 %, respectively. Predictive negative value (PNV) and predictive positive value (PPV) were 94.5 % and 64 % area under the curve (AUC) 0.86. CONCLUSION: Elastosonography provides an interesting contribution to the differentiation of malignant and benign thyroid nodules. Particularly worthy of mention is that an entirely elastic nodule pattern was observed only in relation to benign nodules, a result which would suggest that immediate recourse to FNAB might be avoided.


Subject(s)
Elasticity Imaging Techniques/methods , Image Processing, Computer-Assisted , Thyroid Neoplasms/diagnostic imaging , Thyroid Nodule/diagnostic imaging , Ultrasonography, Doppler, Color/methods , Adenocarcinoma, Follicular/diagnostic imaging , Adenocarcinoma, Follicular/pathology , Adenocarcinoma, Follicular/surgery , Adenocarcinoma, Papillary/diagnostic imaging , Adenocarcinoma, Papillary/pathology , Adenocarcinoma, Papillary/surgery , Adult , Aged , Aged, 80 and over , Biopsy, Fine-Needle , Calcinosis/diagnostic imaging , Diagnosis, Differential , Female , Humans , Male , Middle Aged , Prospective Studies , Sensitivity and Specificity , Thyroid Gland/diagnostic imaging , Thyroid Gland/pathology , Thyroid Neoplasms/pathology , Thyroid Neoplasms/surgery , Thyroid Nodule/pathology , Thyroid Nodule/surgery , Thyroidectomy , Ultrasonography, Interventional , Young Adult
2.
Immunopharmacol Immunotoxicol ; 21(3): 565-82, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10466080

ABSTRACT

CD69, also known as activation inducer molecule, very early activation antigen, MLR-3 and Leu-23, is a member of the natural killer (NK) cell gene complex family of signal transducing receptors. CD69 is as a type II transmembrane glycoprotein with a C-type lectin binding domain in the extracellular portion of the molecule. CD69 expression is induced in vitro on cells of most hematopoietic lineages, including T and B lymphocytes, NK cells, murine macrophages, neutrophils and eosinophils, while it is constitutively expressed on human monocytes, platelets and epidermal Langerhans cells. Although a specific ligand for CD69 has not been identified, its wide cellular distribution and the induction of intracellular signals upon CD69 crosslinking suggest a role for the receptor in the biology of hematopoietic cells. Moreover, certain results indicate that CD69 may be involved in the pathogenesis of such diseases as rheumatoid arthritis, chronic inflammatory liver diseases, mild asthma, and acquired immunodeficiency syndrome.


Subject(s)
Antigens, CD/physiology , Antigens, Differentiation, T-Lymphocyte/physiology , Immunity , Antigens, CD/chemistry , Antigens, Differentiation, T-Lymphocyte/chemistry , Calcium/metabolism , Humans , Lectins, C-Type , Nitric Oxide/biosynthesis , Signal Transduction , T-Lymphocytes/immunology
3.
J Biol Chem ; 274(36): 25411-8, 1999 Sep 03.
Article in English | MEDLINE | ID: mdl-10464270

ABSTRACT

Myristoylated alanine-rich C kinase substrate (MARCKS) and MARCKS-related protein (MRP; MacMARCKS) are protein kinase C substrates in diverse cell types. Activation of murine macrophages by cytokines increases MRP expression, but infection with Leishmania promastigotes during activation results in MRP depletion. We therefore examined the effect of Leishmania major LV39 on recombinant MRP. Both live promastigotes and a soluble fraction of LV39 lysates degraded MRP to yield lower molecular weight fragments. Degradation was independent of MRP myristoylation and was inhibited by protein kinase C-dependent phosphorylation of MRP. MRP was similarly degraded by purified leishmanolysin (gp63), a Leishmania surface metalloprotease. Degradation was evident at low enzyme/substrate ratios, over a broad pH range, and was inhibited by 1,10-phenanthroline and by a hydroxamate dipeptide inhibitor of leishmanolysin. Using mass spectrometric analysis, cleavage was shown to occur within the effector domain of MRP between Ser(92) and Phe(93), in accordance with the substrate specificity of leishmanolysin. Moreover, an MRP construct in which the effector domain had been deleted was resistant to cleavage. Thus, Leishmania infection may result in leishmanolysin-dependent hydrolysis of MRP, a major protein kinase C substrate in macrophages.


Subject(s)
Leishmania major/enzymology , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Amino Acid Sequence , Animals , Hydrolysis , Mass Spectrometry , Metalloendopeptidases/chemistry , Molecular Sequence Data , Protozoan Proteins/metabolism , Substrate Specificity
4.
J Biol Chem ; 274(24): 16782-7, 1999 Jun 11.
Article in English | MEDLINE | ID: mdl-10358020

ABSTRACT

Leishmania, a protozoan parasite of macrophages, has been shown to interfere with host cell signal transduction pathways including protein kinase C (PKC)-dependent signaling. Myristoylated alanine-rich C kinase substrate (MARCKS) and MARCKS-related protein (MRP, MacMARCKS) are PKC substrates in diverse cell types. MARCKS and MRP are thought to regulate the actin network and thereby participate in cellular responses involving cytoskeletal rearrangement. Because MRP is a major PKC substrate in macrophages, we examined its expression in response to infection by Leishmania. Activation of murine macrophages by cytokines increased MRP expression as determined by Western blot analysis. Infection with Leishmania promastigotes at the time of activation or up to 48 h postactivation strongly decreased MRP levels. Leishmania-dependent MRP depletion was confirmed by [3H]myristate labeling and by immunofluorescence microscopy. All species or strains of Leishmania parasites tested, including lipophosphoglycan-deficient Leishmania major L119, decreased MRP levels. MRP depletion was not obtained with other phagocytic stimuli including zymosan, latex beads, or heat-killed Streptococcus mitis, a Gram-positive bacterium. Experiments with [3H]myristate labeled proteins revealed the appearance of lower molecular weight fragments in Leishmania-infected cells suggesting that MRP depletion may be due to proteolytic degradation.


Subject(s)
Bone Marrow Cells/parasitology , Leishmania/pathogenicity , Macrophages/parasitology , Membrane Proteins/biosynthesis , Animals , Calmodulin-Binding Proteins , Cell Compartmentation , Down-Regulation , Interferon-gamma/pharmacology , Intracellular Signaling Peptides and Proteins , Leishmania donovani/pathogenicity , Leishmania major/pathogenicity , Leishmania mexicana/pathogenicity , Macrophage Activation , Macrophages/drug effects , Membrane Proteins/isolation & purification , Mice , Mice, Inbred CBA , Microfilament Proteins , Phagocytosis , Protein Kinase C/metabolism , Species Specificity , Tumor Necrosis Factor-alpha/pharmacology
5.
Shock ; 10(1): 37-42, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9688089

ABSTRACT

Overproduction of nitric oxide (NO) upon expression of inducible NO synthase (iNOS) may be responsible for refractory hypotension in septic shock. Whereas high levels of NOS activity have been documented in experimental models of endotoxemia or intravenous challenge with Escherichia coil, much less is known concerning tissue models of Gram-negative infection. We examined NO production (measured as the accumulation of plasma NO3- + NO2-) in a murine model of Gram-negative peritonitis. Plasma NO3- + NO2- increased progressively from 25 microM to peak levels of 50-150 microM 24 h after intraperitoneal challenge with E. coli 0111:B4, similar to values reported for septic shock patients. Treatment of infected mice with NG-monomethyl-L-arginine, an inhibitor of NOS activity, resulted in the efficient inhibition of NO3- + NO2- production. In order to evaluate the roles of interferon-gamma (IFN-gamma) and tumor necrosis factor (TNF-alpha) in the induction of NO synthesis in murine peritonitis, mice deficient in the respective cytokine receptors were studied. In control in vitro experiments, macrophages from IFN-gammaR- or TNFR55-deficient mice, while failing to respond to IFN-gamma or TNF-alpha, respectively, produced high levels of NO under appropriate stimulation. When challenged intraperitoneally with E. coli, IFN-gammaR- or TNFR55-deficient mice exhibited similar levels of bacteremia and NO production as their wild-type controls. These data thus suggest that enhanced NO production during focal Gram-negative infection may occur in the absence of signaling through either IFN-gammaR or TNFR55.


Subject(s)
Gram-Negative Bacterial Infections/metabolism , Nitric Oxide/metabolism , Peritonitis/metabolism , Receptors, Cytokine/deficiency , Sepsis/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Female , Macrophages/metabolism , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Peritonitis/microbiology , Receptors, Cytokine/genetics , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Interferon gamma Receptor
6.
J Leukoc Biol ; 62(3): 349-55, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9307073

ABSTRACT

CD69, a member of the natural killer cell gene complex family of signal transducing receptors, represents one of the earliest activation antigens in human and murine lymphocytes. In contrast, human monocytes may express CD69 in a constitutive fashion. We have evaluated the expression and function of CD69 in murine bone marrow-derived macrophages. CD69 expression as determined by flow cytometry was not constitutive but was induced by stimulation with interferon-gamma (IFN-gamma) plus bacterial lipopolysaccharide (LPS) or tumor necrosis factor a (TNF-alpha). Stimulation with LPS alone was equally effective. Infection with the protozoan parasite Leishmania did not induce CD69 expression nor influence CD69 up-regulation by IFN-gamma plus LPS. Induction of CD69 expression was significantly inhibited in the presence of prostaglandin E2 or dibutyryl-cAMP. Stimulation of macrophages with anti-CD69 monoclonal antibody in the presence of IFN-gamma induced both nitric oxide production and TNF-alpha release. Moreover, anti-CD69 stimulation of Leishmania-infected macrophages resulted in elimination of the intracellular parasite. These results suggest that CD69 is an activation antigen for murine macrophages and may serve as a signaling receptor for an as yet uncharacterized ligand.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Leishmaniasis/immunology , Macrophage Activation , Macrophages/immunology , Receptors, Immunologic/physiology , Animals , Bone Marrow Cells , Bucladesine/pharmacology , Cytotoxicity, Immunologic , Dinoprostone/pharmacology , Interferon-gamma/pharmacology , Lectins, C-Type , Lipopolysaccharides/pharmacology , Lymphotoxin-alpha/physiology , Mice , Mice, Inbred CBA , Mice, Knockout , Nitric Oxide/pharmacology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects
7.
Crit Care Med ; 25(4): 607-13, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9142024

ABSTRACT

OBJECTIVES: To determine and compare the respective concentrations of tumor necrosis factor (TNF)-alpha, interleukin (IL)-6, soluble TNF receptors, nitrite/nitrate (NO2-/NO3-), and procalcitonin in the plasma of patients with septic shock, cardiogenic shock, and bacterial pneumonia without shock; and to assess the predictive value of these mediators in defining patients with septic shock. DESIGN: Cohort study, comparing normal volunteers (controls) and patients with septic shock, cardiogenic shock, and bacterial pneumonia. SETTING: A collaborative study among an intensive care unit, an emergency room, and three research laboratories. PATIENTS: Mediators were measured at various times in 15 patients with septic shock (during the shock phase and during the recovery phase), in seven patients with cardiogenic shock during the shock phase, and in seven patients with severe bacterial pneumonia on day 1 of admission. INTERVENTIONS: Blood samples were collected at various times during the course of the disease. MEASUREMENTS AND MAIN RESULTS: TNF-alpha values were highest in the acute phase of septic shock (53 to 131 pg/mL during septic shock), while patients with bacterial pneumonia had intermediate concentrations (32 pg/mL). TNF-alpha concentrations were normal in patients with cardiogenic shock. IL-6 concentrations were highest in patients with acute septic shock (85 to 385 pg/mL). However, in contrast to TNF-alpha concentrations, IL-6 concentrations were normal in patients with bacterial pneumonia and increased in patients with cardiogenic shock (78 pg/mL). Soluble TNF receptors were increased in all three groups vs. controls, with the highest increase in patients with septic shock. NO2-/NO3- concentrations were highest (72 to 140 mM) in patients with septic shock, and were < 40 mM in the other groups of patients. Procalcitonin concentrations were only markedly increased in patients with septic shock (72 to 135 ng/mL, compared with approximately 1 ng/mL in the three other groups). The best predictive value for septic shock was found to be the measurements of NO2-/NO3- and procalcitonin concentrations. CONCLUSIONS: These observations showed that increase of proinflammatory cytokines was a consequence of inflammation, not of shock. In this study comparing various shock and infectious states, measurements of NO2-/NO3- concentration and procalcitonin concentration represented the most suitable tests for defining patients with septic shock.


Subject(s)
Calcitonin/blood , Cytokines/blood , Nitrates/blood , Nitrites/blood , Protein Precursors/blood , Receptors, Tumor Necrosis Factor/blood , Shock, Septic/blood , Shock, Septic/diagnosis , Adult , Aged , Aged, 80 and over , Biomarkers , Calcitonin Gene-Related Peptide , Cohort Studies , Diagnosis, Differential , Female , Humans , Interleukin-6/blood , Male , Middle Aged , Pneumonia, Bacterial/blood , Predictive Value of Tests , Shock, Cardiogenic/blood , Tumor Necrosis Factor-alpha/analysis
8.
Int J Cancer ; 62(6): 743-8, 1995 Sep 15.
Article in English | MEDLINE | ID: mdl-7558424

ABSTRACT

Nitric oxide (NO) has been shown to exert cytotoxic effects on tumor cells. We have reported that EC219 cells, a rat-brain-microvessel-derived endothelial cell line, produced NO through cytokine-inducible NO synthase (iNOS), the induction of which was significantly decreased by (a) soluble factor(s) secreted by DHD/PROb, an invasive sub-clone of a rat colon-carcinoma cell line. In this study, the DHD/PROb cell-derived NO-inhibitory factor was characterized. Northern-blot analysis demonstrated that the induction of iNOS mRNA in cytokine-activated EC219 cells was decreased by PROb-cell-conditioned medium. When DHD/PROb cell supernatant was fractionated by affinity chromatography using Con A-Sepharose or heparin-Sepharose, the NO-inhibitory activity was found only in Con A-unbound or heparin-unbound fractions, respectively, indicating that the PROb-derived inhibitory factor was likely to be a non-glycosylated and non-heparin-binding molecule. Pre-incubation of DHD/PROb-cell supernatant with anti-TGF-beta neutralizing antibody completely blocked the DHD/PROb-derived inhibition of NO production by EC219 cells. Addition of exogenous TGF-beta 1 dose-dependently inhibited NO release by EC219 cells. The presence of active TGF-beta in the DHD/PROb cell supernatant was demonstrated using a growth-inhibition assay. Moreover, heat treatment of medium conditioned by the less invasive DHD/REGb cells, which constitutively secreted very low levels of active TGF-beta, increased both TGF-beta activity and the ability to inhibit NO production in EC219 cells. Thus, DHD/PROb colon-carcinoma cells inhibited NO production in EC219 cells by secreting a factor identical or very similar to TGF-beta.


Subject(s)
Brain/blood supply , Colonic Neoplasms/metabolism , Endothelium, Vascular/metabolism , Nitric Oxide/biosynthesis , Transforming Growth Factor beta/physiology , Animals , Cell Communication/physiology , Cell Line , Colonic Neoplasms/pathology , Cytokines/metabolism , Cytokines/physiology , Endothelium, Vascular/cytology , Enzyme Induction , Interferon-gamma/pharmacology , Mice , Microcirculation , Nitric Oxide/physiology , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/metabolism , Rats , Recombinant Proteins , Stimulation, Chemical , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/pharmacology
9.
J Leukoc Biol ; 58(2): 217-24, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7543922

ABSTRACT

The effect of prostaglandin (PG) E2 on macrophage activation by interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF-alpha) was evaluated. Murine macrophages infected with Leishmania enriettii or Leishmania major were activated by exposure to IFN-gamma (10-50 U/ml) and TNF-alpha (30-3000 U/ml), leading to intracellular parasite destruction within 24-48 h. Leishmanicidal activity was markedly increased when activation was performed in the presence of PGE2 (10(-9)-10(-7) M) or arachidonate (10(-5) M, a PG precursor), concomitant with enhanced nitrite release and glucose oxidation through the hexose monophosphate shunt pathway. Conversely, activation was reduced by indomethacin and hydrocortisone, two inhibitors of PG synthesis. Parasite killing and nitrite production were fully restored by exogenous PGE2, indicating that inhibition by these drugs was related to their ability to block PG production. PG can stimulate adenylate cyclase, thus raising intracellular cAMP levels. Accordingly, dibutyryl-cAMP, theophylline (which prevents cAMP breakdown), and forskolin (an activator of adenylate cyclase) all stimulated macrophage activation. Finally, PGE2 and cAMP enhanced expression of inducible nitric oxide synthase mRNA in response to IFN-gamma and TNF-alpha, and this effect was inhibited by the cAMP antagonist 2'-O-methyl adenosine. These findings are consistent with the hypothesis that PGE2 acts as a positive agonist in macrophage activation by IFN-gamma and TNF-alpha via its capacity to modulate intracellular cAMP levels.


Subject(s)
Amino Acid Oxidoreductases/biosynthesis , Bucladesine/pharmacology , Cyclic AMP/metabolism , Dinoprostone/pharmacology , Interferon-gamma/pharmacology , Macrophage Activation/physiology , Tumor Necrosis Factor-alpha/pharmacology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Cell Differentiation , Cells, Cultured , Colforsin/pharmacology , Dose-Response Relationship, Drug , Gene Expression/drug effects , Hematopoietic Stem Cells/cytology , Humans , Kinetics , Leishmania/drug effects , Macrophage Activation/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Nitric Oxide Synthase , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Recombinant Proteins/pharmacology , Theophylline/pharmacology
10.
Int J Cancer ; 59(5): 699-705, 1994 Dec 01.
Article in English | MEDLINE | ID: mdl-7525497

ABSTRACT

Nitric oxide (NO) produced by endothelial cells (EC) has been shown to exert cytotoxic activity on tumor cells. In order to analyze events involved in brain metastasis, the modulation of NO production in rat-brain-derived EC was investigated. NO release was increased by tumor necrosis factor-alpha (TNF-alpha), interferon-gamma (IFN-gamma), interleukin-1 beta, lipopolysaccharide or forskolin in EC219 cells, a rat-brain-microvessel-derived EC line. Dexamethasone decreased NO release by cytokine-activated EC219 cells. Tumor cells (DHD/K12/PROb, a rat colon-carcinoma cell line) were highly adherent to EC219 cells, and adhesion was not modified by TNF-alpha plus IFN-gamma, or by dexamethasone. Addition of tumor cells or tumor-cell-conditioned medium significantly inhibited NO release induced by any of the stimuli examined, but only if added during the initial phase of endothelial-cell activation. Tumor-derived suppression of NO release was also observed in primary cultures of cerebral EC. NO synthase (NOS) activity in cytosol extracts of the cerebral EC line was Ca(2+)-independent and required both NADPH and tetrahydrobiopterin. NOS activity was increased by TNF-alpha and IFN-gamma, and significantly reduced by tumor-cell-conditioned medium. These results suggest that rat colon-carcinoma cells may have developed a protective mechanism involving the release of (a) soluble factor(s) which inhibit(s) NO production by cerebral EC.


Subject(s)
Brain/blood supply , Colonic Neoplasms/metabolism , Cytokines/pharmacology , Endothelium, Vascular/metabolism , Nitric Oxide/biosynthesis , Amino Acid Oxidoreductases/metabolism , Animals , Biopterins/analogs & derivatives , Biopterins/pharmacology , Cell Adhesion , Cell Line , Colforsin/pharmacology , Culture Media, Conditioned , Dexamethasone/pharmacology , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Lipopolysaccharides/pharmacology , NADP/pharmacology , Nitric Oxide Synthase , Rats , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
11.
Biochem J ; 301 ( Pt 1): 243-7, 1994 Jul 01.
Article in English | MEDLINE | ID: mdl-7518670

ABSTRACT

We examined the effect of bafilomycin A1 (BAF), an inhibitor of vacuolar-type H(+)-ATPases, on macrophages activation (measured as increased nitrite production and leishmanicidal activity) induced by interferon gamma alone or together with lipopolysaccharide or tumour necrosis factor alpha. BAF increased intravesicular pH and enhanced nitrite release by activated macrophages; however, the NO concentration necessary to kill parasites was higher in BAF-exposed than control macrophages, suggesting that microbicidal nitrogen derivatives were less active at alkaline pH. Antibody to tumour necrosis factor alpha inhibited BAF-induced nitrite production in interferon-activated cultures. To determine if enhanced NO synthesis was related to vesicular alkalinization, macrophages were incubated with the lysosomotropic bases NH4Cl and methylamine. These agents also increased intravesicular pH and nitrite production. Nitrite production was correlated with enhanced NO synthase activity in cytosolic extracts of the activated cells.


Subject(s)
Leishmania enriettii/immunology , Macrolides , Macrophage Activation/physiology , Nitrites/metabolism , Amino Acid Oxidoreductases/metabolism , Ammonium Chloride/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Cytosol/metabolism , Female , Hydrogen-Ion Concentration , In Vitro Techniques , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophage Activation/immunology , Male , Methylamines/pharmacology , Mice , Mice, Inbred CBA , Nigericin/pharmacology , Nitric Oxide Synthase , Proton-Translocating ATPases/antagonists & inhibitors , Recombinant Proteins , Tumor Necrosis Factor-alpha/pharmacology
12.
J Infect Dis ; 169(1): 105-11, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8277172

ABSTRACT

A recombinant (r) NH2-terminal fragment of bactericidal/permeability-increasing protein, rBPI23, was shown to inhibit murine macrophage nitric oxide (NO) production elicited by lipopolysaccharide (LPS) plus interferon-gamma (IFN-gamma). Normal mouse plasma amplified NO synthesis (measured as NO2- release) at LPS concentrations of 1-10 ng/mL, and antibody to the plasma LPS-binding protein (LBP) partially inhibited NO2- release in the presence of normal mouse plasma. rBPI23 (1 microgram/mL) effectively inhibited LPS-dependent NO2- release in the presence or absence of normal mouse plasma. Fifty percent inhibition of IFN-gamma/LPS-elicited NO2- production or of binding of fluoresceinated LPS was obtained with approximately 0.2 microgram/mL rBPI23. These results provide a basis for studies of rBPI23 effects on NO synthase activity in murine models of gram-negative sepsis.


Subject(s)
Blood Bactericidal Activity/drug effects , Blood Proteins/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Membrane Proteins , Nitric Oxide/biosynthesis , Animals , Antimicrobial Cationic Peptides , Cells, Cultured , Dose-Response Relationship, Drug , Female , Interferon-gamma/pharmacology , Leishmania enriettii/physiology , Lipopolysaccharides/metabolism , Macrophage Activation , Macrophages/drug effects , Macrophages/immunology , Macrophages/parasitology , Mice , Mice, Inbred CBA , Permeability , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis
13.
J Leukoc Biol ; 54(5): 423-9, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8228620

ABSTRACT

We have examined the effects of transforming growth factor beta 1 (TGF-beta 1) on the regulation of murine bone marrow-derived macrophage function. TGF-beta, added simultaneously with or up to 4 h before interferon-gamma (IFN-gamma) plus lipopolysaccharide (LPS), inhibited macrophage leishmanicidal activity, nitrite (NO2-) production, and secretion of prostaglandin E2. In contrast, no effect of TGF-beta could be demonstrated on macrophages stimulated with IFN-gamma plus tumor necrosis factor-alpha (TNF-alpha) under the same conditions. These results suggested that TGF-beta inhibited LPS-induced triggering of macrophage activation, which was confirmed by studies with IFN-gamma-primed cells. Interestingly, when macrophages were pretreated with TGF-beta for 24 h, NO2- production in response to IFN-gamma plus TNF-alpha was also inhibited. Although control and IFN-gamma/LPS-stimulated macrophages were found to secrete latent TGF-beta, only the IFN-gamma/LPS cultures produced biologically active TGF-beta. Significantly, active TGF-beta was present at concentrations shown earlier to inhibit macrophage function.


Subject(s)
Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophage Activation/physiology , Transforming Growth Factor beta/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Bone Marrow/physiology , Bone Marrow Cells , Cells, Cultured , Dinoprostone/metabolism , Leishmania enriettii/metabolism , Macrophage Activation/drug effects , Macrophages/cytology , Macrophages/metabolism , Macrophages/physiology , Mice , Mice, Inbred CBA , Nitrites/metabolism
14.
Eur J Immunol ; 23(8): 2045-8, 1993 Aug.
Article in English | MEDLINE | ID: mdl-7688311

ABSTRACT

Interleukin-10 (IL-10) has been reported to inhibit nitric oxide (NO) synthesis and microbicidal activity of interferon-gamma (IFN-gamma)-stimulated macrophages (M phi) by preventing the secretion of tumor necrosis factor-alpha (TNF-alpha) which serves as an autocrine activating signal. We have examined the effects of recombinant IL-10 on the capacity of IFN-gamma together with exogenous TNF-alpha to induce NO synthesis by bone marrow-derived M phi. Under these conditions and in contrast to its reported deactivating potential, IL-10 strongly enhanced NO synthesis measured as nitrite (NO2-) release (half maximal stimulation at approximately 10 U/ml). IL-10 further increased NO2- production by M phi stimulated in the presence of optimal concentrations of prostaglandin E2, a positive modulator of M phi activation by IFN-gamma/TNF-alpha. Increased steady state levels of NO synthase mRNA were observed in 4-h IFN-gamma/TNF-alpha cultures and enhanced NO2(-)-release was evident 24 h but not 48 h after stimulation. These results suggest that the effects of IL-10 on M phi function are more complex than previously recognized.


Subject(s)
Interferon-gamma/pharmacology , Interleukin-10/pharmacology , Macrophages/metabolism , Nitric Oxide/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Amino Acid Oxidoreductases/genetics , Animals , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C3H , Mice, Inbred CBA , Nitric Oxide Synthase , Nitrites/metabolism , RNA, Messenger/analysis , Recombinant Proteins/pharmacology
15.
J Infect Dis ; 167(6): 1351-7, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8501324

ABSTRACT

The bactericidal/permeability-increasing protein (BPI) inhibits the lipopolysaccharide (LPS)-mediated activation of monocytes. Due to its inhibitory activity for various LPS, BPI has therapeutic potential in endotoxic shock. To be efficient in vivo, BPI should overcome the action of LPS-binding protein (LBP), a serum molecule that increases the expression of LPS-inducible genes via CD14 of monocytes, rBPI23, a recombinant fragment of BPI, prevented in a dose-dependent manner the binding and the internalization of LPS mediated by LBP. Consequently, rBPI23 also inhibited LPS-induced tumor necrosis factor (TNF alpha) synthesis from monocytes. LPS- and LBP-mediated activation of monocytes was totally inhibited when LPS was preincubated with rBPI23. Adding rBPI23 at the same time as LBP resulted in an important but partial inhibition of TNF alpha release, but this inhibition vanished with delaying the time of addition of rBPI23. These studies suggest that the inhibitory activity of BPI is related to its ability to compete with LBP for LPS.


Subject(s)
Acute-Phase Proteins , Blood Proteins/metabolism , Carrier Proteins/metabolism , Lipopolysaccharides/metabolism , Membrane Glycoproteins , Membrane Proteins , Monocytes/metabolism , Animals , Antimicrobial Cationic Peptides , Binding, Competitive , Fluorescein-5-isothiocyanate , Humans , In Vitro Techniques , Lipopolysaccharides/antagonists & inhibitors , Mice , Microscopy, Fluorescence , Tumor Necrosis Factor-alpha/metabolism
16.
Glia ; 7(3): 255-62, 1993 Mar.
Article in English | MEDLINE | ID: mdl-7681038

ABSTRACT

Nitric oxide (NO) is a short-lived diffusable molecule now believed to participate in multiple physiologic functions in the CNS including neurotransmission and the maintenance of vascular tone. Previously, we reported that cell lines obtained by retroviral immortalization of tissue macrophages (M phi) could be induced to synthesize nitrite (NO2-), a stable end product of the NO synthetic pathway. We have further characterized the induction and activity of this pathway in a panel of seven microglial clones derived from primary embryonic mouse brain cultures. Like M phi, these clones were found to release high levels of NO2- in response to recombinant interferon-gamma (rIFN-gamma) as a priming signal together with either bacterial lipopolysaccharide (LPS) or exogenous recombinant tumor necrosis factor-alpha (rTNF-alpha). As previously demonstrated for M phi, phagocytosis of zymosan particles during induction of enzyme activity enhanced subsequent NO2- production, which is of interest in light of the postulated phagocytic role of microglia within the CNS. Biochemical characterization of enzyme activity in intact microglial clones and in isolated cytosolic fractions indicates that the microglial NO synthase present in these murine cell clones represents the M phi-like isotype. These findings suggest that microglial cells could represent a major source of NO within the CNS.


Subject(s)
Amino Acid Oxidoreductases/biosynthesis , Neuroglia/enzymology , Nitric Oxide/metabolism , Animals , Brain/cytology , Brain/embryology , Enzyme Induction/drug effects , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/physiology , Mice , Mice, Inbred CBA , Neuroglia/drug effects , Nitric Oxide Synthase , Phagocytosis , Recombinant Fusion Proteins/pharmacology , Recombinant Proteins , Tumor Necrosis Factor-alpha/pharmacology , Zymosan/pharmacology
17.
J Leukoc Biol ; 52(4): 363-8, 1992 Oct.
Article in English | MEDLINE | ID: mdl-1402386

ABSTRACT

We have studied the effects of highly purified rabbit lipopolysaccharide (LPS)-binding protein (LBP) on the ability of murine bone marrow-derived macrophages to respond to bacterial LPS. Macrophage responses studied include the secretion of tumor necrosis factor alpha, production of arginine-derived nitrite (NO2-), and killing of an intracellular pathogen, Leishmania enriettii. Macrophages from either CBA or LPS-hyporesponsive C3H/HeJ mice exhibited significantly greater sensitivity to LPS in the presence of LBP. Furthermore, both CBA and C3H/HeJ macrophages demonstrated an LBP-dependent enhancement of LPS binding. These results suggest that C3H/HeJ macrophages are capable of binding LPS-LBP complexes and support the hypothesis that hyporesponsiveness in this strain involves a step subsequent to LPS binding. Furthermore, these findings provide additional evidence of the important role played by the acute-phase plasma protein LBP in modifying host response to LPS.


Subject(s)
Acute-Phase Proteins , Carrier Proteins/pharmacology , Lipopolysaccharides , Macrophages/metabolism , Membrane Glycoproteins , Animals , Cell Separation/methods , Cells, Cultured , Flow Cytometry , Leishmania/drug effects , Lipopolysaccharides/metabolism , Macrophage Activation/drug effects , Macrophages/drug effects , Mice , Mice, Inbred C3H , Mice, Inbred CBA , Nitrites/metabolism , Nitrogen Dioxide/metabolism , Sensitivity and Specificity , Tumor Necrosis Factor-alpha/metabolism
18.
Biochem J ; 284 ( Pt 2): 387-92, 1992 Jun 01.
Article in English | MEDLINE | ID: mdl-1599422

ABSTRACT

Mouse macrophages activated by interferon-gamma kill intracellular Leishmania by a process that depends on the generation of L-arginine-derived nitrogen oxidation products. Interferon-induced intracellular killing can be mimicked by exposure of macrophages to the Ca2+ ionophore A23187 in the presence of lipopolysaccharide. The mechanisms of this effect were therefore investigated. Destruction of the parasite was accompanied by accumulation of nitrite in the macrophage culture fluids. Leishmanicidal activity and nitrite production in cultures stimulated with ionophore A23187 and lipopolysaccharide were abrogated when cells were activated in medium containing arginase or the L-arginine analogues L-canavanine, guanidine or NG-monomethyl-L-arginine. L-Arginine was required during the lipopolysaccharide-induced triggering phase only. Indeed, macrophage priming with ionophore A23187 in L-arginine-depleted medium led to full microbicidal activity and nitrite generation provided that L-arginine was present during subsequent triggering by lipopolysaccharide. Addition of NG-monomethyl-L-arginine to ionophore-activated macrophages increased O2- production on phorbol myristate stimulation, while inhibiting glucose oxidation through the hexose monophosphate shunt pathway. Leishmanicidal activity and nitrite production were also inhibited when ionophore-treated cultures were incubated with excess iron, implying a role for iron as a defence mechanism against the toxicity of nitrogen derivatives. These results indicate that the ionophore-induced leishmanicidal activity occurs through a process similar to that evoked by interferon-gamma, i.e. the production of L-arginine-derived nitrogen oxidation products.


Subject(s)
Calcimycin/pharmacology , Leishmania mexicana/immunology , Macrophage Activation , Macrophages/immunology , Nitrogen/metabolism , Animals , Arginine/metabolism , Bone Marrow Cells , Cells, Cultured , Ferrous Compounds/pharmacology , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Oxidation-Reduction
19.
J Immunol ; 148(4): 1171-5, 1992 Feb 15.
Article in English | MEDLINE | ID: mdl-1310709

ABSTRACT

Calcium ionophore A23187 can mimic IFN-gamma-induced macrophage activation for intracellular Leishmania killing and secretion of L-arginine-derived nitrite. Because the effects of ionophore are not restricted to calcium mobilization but also involve alterations of phospholipid metabolism, we have examined the role of PGE2 in the activation process. Macrophages exposed to A23187 or IFN-gamma in the presence of LPS and FCS secreted significant amounts of PGE2 independently of the presence of L-arginine in the incubation medium. The addition of the cyclooxygenase inhibitor indomethacin or omission of FCS abrogated PGE2 secretion but had little effect on nitrite production or intracellular killing. The addition of exogenous PGE2, of agents increasing PGE2 production such as arachidonic acid and colchicine, or of an analogue of cAMP, dibutyryl cAMP inhibited A23187 + LPS-induced activation whereas that mediated by IFN-gamma + LPS remained unimpaired. Our results indicate that PGE2 can modulate activation induced by A23187 but not by IFN-gamma, probably by a process involving cAMP. Conceivably, ionophore can mimic IFN-gamma for the induction of activation but lacks the capacity to help maintain the activated state because of its inability to desensitize macrophages to negative regulation by PGE2, as suggested previously for IFN-gamma-dependent activation.


Subject(s)
Calcimycin/pharmacology , Dinoprostone/pharmacology , Interferon-gamma/pharmacology , Macrophage Activation/drug effects , Animals , Cyclic AMP/physiology , Dinoprostone/biosynthesis , Female , Interferon-alpha/pharmacology , Leishmania/immunology , Lipopolysaccharides , Male , Mice , Mice, Inbred CBA , Nitrites/metabolism
20.
Biochem Biophys Res Commun ; 183(1): 150-5, 1992 Feb 28.
Article in English | MEDLINE | ID: mdl-1371917

ABSTRACT

Murine macrophages activated by interferon-gamma and lipopolysaccharide become leishmanicidal through a process involving L-arginine-derived nitrogen oxidation products. Both nitrite secretion and parasite killing by activated macrophages were inhibited by 3-amino-1,2,4-triazole as well as the related compound, 3-amino-1,2,4-triazine. Moreover, NO synthase activity in cytosolic extracts of activated cells was inhibited by both compounds. 4-amino-1,2,4-triazole, an isomer of 3-amino-1,2,4-triazole, was without effect. Our results suggest that besides its known inhibitory effect on catalases and peroxidases, 3-amino-1,2,4-triazole is an inhibitor of NO synthase. The resemblance between the tautomeric form of 3-amino-1,2,4-triazole and the guanidino group of L-arginine, the natural substrate for NO synthase, might be responsible for the observed inhibition.


Subject(s)
Amino Acid Oxidoreductases/antagonists & inhibitors , Amitrole/pharmacology , Leishmania/drug effects , Macrophage Activation/physiology , Macrophages/enzymology , Nitric Oxide/metabolism , Animals , Cytosol/enzymology , Dose-Response Relationship, Drug , Interferon-gamma/pharmacology , Lipopolysaccharides/metabolism , Macrophage Activation/drug effects , Macrophages/drug effects , Mice , Mice, Inbred CBA , Nitric Oxide Synthase
SELECTION OF CITATIONS
SEARCH DETAIL
...