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











Publication year range
1.
Biomedica ; 36(2): 239-50, 2016 Jun 03.
Article in English | MEDLINE | ID: mdl-27622485

ABSTRACT

INTRODUCTION: Vitamin D3 (VD3) has been described as a modulator of immune system cells, including dendritic cells (DC). Previous studies have shown its importance in in vitro generation of tolerogenic DC, which have a similar function and phenotype to that of CD141 dermal DCs that produce IL-10 and induce (LTreg) CD4+ T regulator cells.  OBJECTIVE: This paper presents a study that compares the phenotype and cytokines produced by DC generated in presence and absence of VD3, which were matured with lipopolysaccharide (LPS), and their ability to induce LTreg from naïve allogeneic CD4+ T cells.  MATERIALS AND METHODS: In order to compare them, peripheral blood mononuclear cells were isolated to select monocytes CD14+ T cells and differentiate them in vitro from DC in the presence and absence of VD3, and to mature them with LPS. Phenotype and cytokine levels were also analyzed in the culture supernatants. Dendritic cells were then co-cultured with naïve allogeneic CD4+ T cells and the frequencies of LTreg were determined (naïve-activated).  RESULTS: The results showed that unstimulated DC generated with VD3 kept the CD14. When activated with LPS, they expressed lower levels of C83, CD83 and CD86; HLA-DR; higher amounts of IL-1ß, IL-8, IL-10, and tended to lessen IL-6, IL-12p70 and TGF-ß1, compared to DCs not treated with VD3. The frequency of naïve LTreg was similar, although immature DC generated with VD3 tended to induce activated LTregs.  CONCLUSION: Based on these results, it is possible to conclude that DCs generated with VD3 and treated with LPS presented a 'semi-mature' phenotype, and were able to secrete pro-inflammatory and anti-inflammatory cytokines. Besides, they did not increase their capacity to promote the polarization of naïve allogenic CD4+ T cells towards LTregs.


Subject(s)
Cholecalciferol/pharmacology , Cytokines/biosynthesis , Cytokines/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Interleukin-10/metabolism , Interleukin-8/metabolism , Leukocytes, Mononuclear/immunology , Lipopolysaccharides/chemistry , Monocytes/drug effects , Monocytes/immunology , T-Lymphocytes, Regulatory/immunology , Cells, Cultured , Cholecalciferol/metabolism , Dendritic Cells/cytology , Humans , Interleukin-10/immunology , Interleukin-10/physiology , Interleukin-8/immunology , Interleukin-8/physiology , Leukocytes, Mononuclear/chemistry , Leukocytes, Mononuclear/physiology , Lipopolysaccharides/metabolism , T-Lymphocytes, Regulatory/physiology
2.
J Periodontol ; 87(8): 990-6, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27063996

ABSTRACT

BACKGROUND: Multiple factors affect oral mucosal healing, such as the persistence of an inflammatory reaction. The present study evaluates effects of tumor necrosis factor (TNF)-α and interleukin (IL)-1ß, IL-6, and IL-8 on epithelial cells (ECs) and human gingival fibroblasts (GFs) in vitro. METHODS: GFs and ECs were seeded in 96-well plates (1 × 10(4) cells/well) in plain culture medium (Dulbecco's modified Eagle's medium [DMEM]) containing 1% antibiotic/antimycotic solution and 10% fetal bovine serum, and incubated for 24 hours. Both cell lines were exposed for 24 hours to the following cytokines: 1) TNF-α (100 ng/mL); 2) IL-1ß (1 ng/mL); 3) IL-6 (10 ng/mL); and 4) IL-8 (10 ng/mL). All cytokines were diluted in serum-free DMEM. Control cultures were exposed only to serum-free DMEM. Effects of exposure to inflammatory cytokines were determined by means of: 1) apoptosis (anexin V); 2) cell migration (wound healing assay); 3) inflammatory cytokine synthesis (enzyme-linked immunosorbent assay). Data were analyzed by Kruskal-Wallis and Mann-Whitney U tests (α = 0.05). RESULTS: Increased apoptosis rates were noted when cells were exposed to inflammatory cytokines, except ECs exposed to IL-1ß. Cell migration was negatively affected by all inflammatory cytokines for both cell lines. ECs and GFs exposed to IL-6 and IL-8 significantly increased synthesis of TNF-α and IL-1ß. CONCLUSIONS: Demonstrated results indicate negative effects of tested inflammatory cytokines on ECs and GFs, inducing apoptosis and impairing cell migration. These results can justify delayed oral mucosa healing in the presence of inflammatory reaction.


Subject(s)
Apoptosis , Fibroblasts/physiology , Interleukin-1beta/physiology , Interleukin-6/physiology , Interleukin-8/physiology , Tumor Necrosis Factor-alpha/physiology , Wound Healing , Cells, Cultured , Epithelial Cells , Gingiva/cytology , Gingiva/metabolism , Humans , Interleukins
3.
Clin Exp Immunol ; 167(1): 129-36, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22132892

ABSTRACT

The disturbed cytokine-chemokine network could play an important role in the onset of diseases with inflammatory processes such as chronic idiopathic urticaria (CIU). Our main objectives were to evaluate the relation between proinflammatory chemokine serum levels from CIU patients and their response to autologous skin test (ASST) and basophil histamine release (BHR). We also aimed to assess the chemokine secretion by peripheral blood mononuclear cells (PBMC) upon polyclonal stimulus and to evaluate chemokine C-C ligand 2/C-X-C chemokine 8 (CCL2/CXCL8) and Toll-like receptor-4 (TLR-4) expression in monocytes. We observed significantly higher serum levels of the CXCL8, CXCL9, CXCL10 and CCL2 in CIU patients compared to the healthy group, regardless of the BHR or ASST response. The basal secretion of CCL2 by PBMC or induced by Staphylococcus aureus enterotoxin A (SEA) was higher in CIU patients than in the control group, as well as for CXCL8 and CCL5 secretions upon phytohaemagglutinin stimulation. Also, up-regulation of CCL2 and CXCL8 mRNA expression was found in monocytes of patients upon SEA stimulation. The findings showed a high responsiveness of monocytes through CCL2/CXCL8 expression, contributing to the creation of a proinflammatory environment in CIU.


Subject(s)
Chemokine CCL2/biosynthesis , Interleukin-8/biosynthesis , Leukocytes, Mononuclear/metabolism , Monocytes/metabolism , Urticaria/genetics , Adult , Aged , Aged, 80 and over , Basophil Degranulation Test , Chemokine CCL2/genetics , Chemokine CCL2/physiology , Chemokines/blood , Chronic Disease , Enterotoxins/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Inflammation , Interleukin-8/genetics , Interleukin-8/physiology , Leukocytes, Mononuclear/drug effects , Lymphocyte Activation/drug effects , Male , Middle Aged , Monocytes/drug effects , Phytohemagglutinins/pharmacology , Real-Time Polymerase Chain Reaction , Skin Tests , Toll-Like Receptor 4/biosynthesis , Toll-Like Receptor 4/genetics , Up-Regulation/drug effects , Urticaria/blood , Urticaria/immunology , Young Adult
4.
Toxicol Appl Pharmacol ; 258(1): 10-8, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22015448

ABSTRACT

The association between chronic human exposure to arsenicals and bladder cancer development is well recognized; however, the underlying molecular mechanisms have not been fully determined. We propose that inflammatory responses can play a pathogenic role in arsenic-related bladder carcinogenesis. In previous studies, it was demonstrated that chronic exposure to 50 nM monomethylarsenous acid [MMA(III)] leads to malignant transformation of an immortalized model of urothelial cells (UROtsa), with only 3 mo of exposure necessary to trigger the transformation-related changes. In the three-month window of exposure, the cells over-expressed pro-inflammatory cytokines (IL-1ß, IL-6 and IL-8), consistent with the sustained activation of NFKß and AP1/c-jun, ERK2, and STAT3. IL-8 was over-expressed within hours after exposure to MMA(III), and sustained over-expression was observed during chronic exposure. In this study, we profiled IL-8 expression in UROtsa cells exposed to 50 nM MMA(III) for 1 to 5 mo. IL-8 expression was increased mainly in cells after 3 mo MMA(III) exposure, and its production was also found increased in tumors derived from these cells after heterotransplantation in SCID mice. UROtsa cells do express both receptors, CXCR1 and CXCR2, suggesting that autocrine cell activation could be important in cell transformation. Supporting this observation and consistent with IL-8 over-expression, CXCR1 internalization was significantly increased after three months of exposure to MMA(III). The expression of MMP-9, cyclin D1, bcl-2, and VGEF was significantly increased in cells exposed to MMA(III) for 3 mo, but these mitogen-activated kinases were significantly decreased after IL-8 gene silencing, together with a decrease in cell proliferation rate and in anchorage-independent colony formation. These results suggest a relevant role of IL-8 in MMA(III)-induced UROtsa cell transformation.


Subject(s)
Cell Transformation, Neoplastic/chemically induced , Interleukin-8/physiology , Organometallic Compounds/toxicity , Urinary Bladder Neoplasms/chemically induced , Animals , Cell Proliferation , Cells, Cultured , Humans , Male , Mice , Mice, SCID , Neoplasm Invasiveness , Receptors, Interleukin-8A/analysis , Receptors, Interleukin-8B/analysis , Urinary Bladder Neoplasms/pathology , Urothelium
5.
Pediatr Blood Cancer ; 56(4): 568-77, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21298741

ABSTRACT

BACKGROUND: The interactions of acute lymphoblastic leukemia (ALL) blasts with bone marrow (BM) stromal cells have a positive impact on leukemia cell survival. In the present study, we proposed to identify and investigate the role of molecules critically involved in leukemia--microenvironment crosstalk. PROCEDURE: Gene expression profiling analyses of BM mesenchymal stem cells (BMMSC) were performed following stimulation by ALL cells. CCL2 and IL-8 plasma levels were evaluated from ALL patients and controls. Expression of the CCL2 and IL-8 receptors in ALL was determined by RT-PCR. The biological effects of CCL2, IL-8 or its neutralizing antibodies in primary precursor-B ALL and BMMSC cells were evaluated using in vitro assays. RESULTS: Leukemia stimulation of BMMSC upregulated the expression of several inflammatory chemokines, including CCL2 and IL-8. The BM plasma levels of CCL2 and IL-8 in children at diagnosis were significantly higher than in healthy controls (P < 0.001). Functional studies revealed that CCL2 and IL-8 enhanced the capacity of BMMSC to support adhesion of ALL cells. CCL2 and IL-8 were also found to enhance BMMSC survival and to increase their proliferation. ALL cells were not directly affected by CCL2 or IL-8. CONCLUSIONS: The leukemic BM microenvironment had increased levels of CCL2 and IL-8. These chemokines are known to have suppressive effects in normal hematopoiesis. Our data indicate that CCL2 and IL-8 have a positive impact on BMMSC survival, proliferation, and adhesiveness to ALL cells. Leukemia-associated CCL2 and IL-8 upregulation may represent one possible mechanism of microenvironment perversion in favor of ALL cells.


Subject(s)
Bone Marrow Cells/metabolism , Chemokine CCL2/metabolism , Interleukin-8/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Cell Adhesion , Cell Survival , Chemokine CCL2/genetics , Chemokine CCL2/physiology , Child , Child, Preschool , Female , Humans , Infant , Interleukin-8/genetics , Interleukin-8/physiology , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Prognosis , Signal Transduction
6.
Scand J Immunol ; 69(2): 73-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19144080

ABSTRACT

Paracoccidioidomycosis (PCM) is a systemic mycosis caused by Paracoccidiodes brasiliensis that presents a wide spectrum of clinical manifestations. Because of the great number of neutrophils polymorphonuclear neutrophils (PMN) found in the P. brasiliensis granuloma, studies have been done to evaluate the role of these cells during the development of the infection. This fungus is found intracellularly in PMN and monocytes/macrophages, suggesting that it is capable of evading damage and surviving inside these cells. Thus, in the present study, we investigated whether P. brasiliensis can prolong the lifetime of PMN, and if this process would be related with IL-8 levels. PMN apoptosis and intracellular levels of IL-8 were analysed by flow cytometry and culture supernatants IL-8 levels were evaluated by enzyme-linked immunosorbent assay. We found that coincubation with P. brasiliensis yeast cells results in an inhibition of PMN apoptosis, which was associated with increase in IL-8 production by these cells. Cocultures treatment with monoclonal antibody anti-IL-8 reversed the inhibitory effect of P. brasiliensis on PMN apoptosis, besides to increase spontaneous apoptosis of these cells. These data show that, in contrast to other microbial pathogens that drive phagocytes into apoptosis to escape killing, P. brasiliensis can extend the lifetime of normal human PMN by inducing autocrine IL-8 production.


Subject(s)
Apoptosis , Interleukin-8/physiology , Neutrophils/physiology , Paracoccidioides/physiology , Adult , Antibodies, Monoclonal/immunology , Humans , Interleukin-8/metabolism , Middle Aged , Phagocytosis
7.
Inflamm Res ; 56(9): 368-76, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17878999

ABSTRACT

OBJECTIVE AND DESIGN: The Macrophage-derived Neutrophil Chemotactic Factor (MNCF) has been characterized as a dexamethasone-resistant neutrophil chemotactic lectin produced by rat macrophages. This study was undertaken to evaluate different MNCF cellular sources and investigate the mechanisms by which MNCF overcomes the anti-inflammatory actions of dexamethasone. MATERIAL AND METHODS: The mouse macrophage-like cell line P388D1 and thioglycollate-elicited mouse macrophages were studied regarding their capacity to release MNCF. Neutrophil migration assays were performed in vivo and in vitro, in either the presence or absence of extracellular matrix glycoproteins (ECM). RESULTS: Mouse and P388D1 macrophages release a lectin that reproduces the activities of rat MNCF. The ability of MNCF to induce neutrophil adhesion and haptotaxis is enhanced through its interaction with laminin and fibronectin. These properties are not inhibited by dexamethasone. CONCLUSIONS: Together, our results suggest that dexamethasone-resistant neutrophil migration induced by MNCF occurs probably because of its interactions with ECM.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dexamethasone/pharmacology , Fibronectins/physiology , Interleukin-8/physiology , Laminin/physiology , Neutrophils/physiology , Animals , Cell Line , Cell Movement , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred BALB C , Tumor Necrosis Factor-alpha/physiology
8.
Toxicon ; 40(12): 1709-17, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12457883

ABSTRACT

Staphylococcus aureus secretes enterotoxins which are superantigens and the major cause of food poisoning in man. Staphylococcal enterotoxins types A and B can induce neutrophil migration into the peritoneal cavity of mice through sensory C-fiber neuropeptides, lipoxygenase or cyclooxygenase metabolites, nitric oxide, histamine, platelet-activating factor and resident macrophages. In this work, we examined the influence of macrophage-derived products on neutrophil migration during peritonitis induced by staphylococcal enterotoxin type B (SEB) in mice. Macrophages stimulated with SEB released a thermolabile neutrophil chemotactic protein with a molecular weight of 1,000-3,000 (by ultrafiltration). This release was inhibited 30% by dexamethasone (an inhibitor of cytokine synthesis and phospholipase A(2) activity), but not by indomethacin (a cyclooxygenase inhibitor) or BW755C (a dual cyclo- and lipoxygenase inhibitor). Dexamethasone also inhibited (100%) the neutrophil migration induced by the chemotactic protein. Similar inhibition occurred in mice pretreated with BWA4C (lipoxygenase inhibitor; 90%), BW755C (99%), BN52021 (platelet-activating factor-acether receptor antagonist; 93%), cimetidine (histamine H(2) receptor antagonist; 76%), capsaicin (a depletor of sensory C-fiber neuropeptides; 82%) and the neurokinin-1 receptor antagonist SR140333 (71%), but not by indomethacin or the neurokinin(2) receptor antagonist SR48968. These results confirm that macrophages are involved in the neutrophil recruitment induced by SEB, and that the chemotactic protein apparently induces neutrophil migration by a mechanism mediated by platelet-activating factor, histamine H(2) receptors, lipoxygenase products and substance P.


Subject(s)
Benzeneacetamides , Chemotaxis, Leukocyte/drug effects , Diterpenes , Enterotoxins/pharmacology , Interleukin-8/physiology , Neutrophil Infiltration/drug effects , 4,5-Dihydro-1-(3-(trifluoromethyl)phenyl)-1H-pyrazol-3-amine/pharmacology , Animals , Capsaicin/pharmacology , Chemotaxis, Leukocyte/physiology , Cimetidine/pharmacology , Dexamethasone/pharmacology , Enzyme Inhibitors/pharmacology , Ginkgolides , Hydroxamic Acids/pharmacology , Indomethacin/pharmacology , Lactones/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Neutrophil Infiltration/physiology , Piperidines/pharmacology , Quinuclidines/pharmacology , Rats , Rats, Wistar
9.
Inflamm Res ; 51(3): 144-53, 2002 Mar.
Article in English | MEDLINE | ID: mdl-12005205

ABSTRACT

OBJECTIVE AND DESIGN: In this study we investigated the chemotactic mediators involved in the Sephadex-induced eosinophil migration into the peritoneal cavities of rats and mice, and which resident peritoneal cells release these mediators. MATERIALS AND METHODS: Sephadex suspension was injected into the peritoneal cavities of rats or mice which were pretreated, or not, with specific drugs that inhibit synthesis or production of the inflammatory mediators and eosinophil chemotactic activities were observed. To investigate the role of resident peritoneal cells as a source of these chemotactic factors, the macrophage population was enhanced or the mast cell population was depleted. The resident cells were also stimulated, in vitro, with Sephadex and the chemotactic activity of the supernatants was determined. RESULTS: Sephadex induced dose and time dependent eosinophil migration in rats and mouse, which were inhibited by dexamethasone and MK 886. BN 52021 only affected the eosinophil migration into the mouse peritoneal cavity. An increase in the macrophage population did not alter the eosinophil migration induced by Sephadex in rat or mouse. However, mast cell population depletion reduced eosinophil migration in rats, but did not alter the migration in mice. Sephadex-stimulated rat mast cells released an eosinophil chemotactic factor whose release was inhibited by dexamethasone and MK 886. Anti-TNF-alpha and anti-IL-8 Abs inhibited the chemotactic activity of the mast cell supernatant. CONCLUSION: Sephadex-induced eosinophil migration into the rat peritoneal cavity is dependent on mast cells, which release LTB4, TNF-alpha and CINC-1. Conversely, Sephadex-induced eosinophil migration into the mouse peritoneal cavity is mediated by PAF and LTB4, which are not released from resident macrophages or mast cells.


Subject(s)
Chemokines, CXC , Dextrans/pharmacology , Eosinophils/drug effects , Interleukin-8/physiology , Leukotriene B4/physiology , Mast Cells/physiology , Peritoneal Cavity/cytology , Platelet Activating Factor/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Cell Movement/drug effects , Chemokine CXCL1 , Chemokines/physiology , Chemotactic Factors/physiology , Dose-Response Relationship, Drug , Eosinophils/physiology , Intercellular Signaling Peptides and Proteins/physiology , Male , Mice , Rats , Rats, Wistar
10.
Mem Inst Oswaldo Cruz ; 92 Suppl 2: 205-10, 1997.
Article in English | MEDLINE | ID: mdl-9698935

ABSTRACT

There are several experimental models describing in vivo eosinophil (EO) migration, including ip injection of a large volume of saline (SAL) or Sephadex beads (SEP). The aim of this study was to investigate the mechanisms involved in the EO migration in these two models. Two consecutive injections of SAL given 48 hr apart, induced a selective recruitment of EO into peritoneal cavity of rats, which peaked 48 hr after the last injection. SEP, when injected ip, promoted EO accumulation in rats. The phenomenon was dose-related and peaked 48 hr after SEP injection. To investigate the mediators involved in this process we showed that BW A4C, MK 886 and dexamethasone (DXA) inhibited the EO migration induced by SAL and SEP. To investigate the source of the EO chemotactic factor we showed that mast cells, macrophages (MO), but not lymphocytes, incubated in vitro in presence of SAL released a factor which induced EO migration. With SEP, only mast cells release a factor that induced EO migration, which was inhibited by BW A4c, MK 886 and DXA. Furthermore, the chemotactic activity of SAL-stimulated mast cells was inhibited by antisera against IL-5 and IL-8 (interleukins). SAL-stimulated MO were only inhibited by anti-IL-8 antibodies as well SEP-stimulated mast cells. These results suggest that the EO migration induced by SAL may be dependent on resident mast cells and MO and mediated by LTB4, IL-5 and IL-8. SEP-induced EO migration was dependent on mast cells and may be mediated by LTB4 and IL-8. Furthermore, IL-5 and IL-8 induced EO migration, which was also dependent on resident cells and mediated by LTB4. In conclusion, EO migration induced by SAL is dependent on mast cells and MO, whereas that induced by SEP is dependent on mast cells alone. Stimulated mast cells release LTB4, IL-5 and IL-8 while MO release LTB4 and IL-8. The IL-5 and IL-8 release by the SAL or SEP-stimulated resident cells may act in an autocrine fashion, thus potentiating LTB4 release.


Subject(s)
Cell Movement/physiology , Eosinophils/physiology , Interleukin-5/physiology , Interleukin-8/physiology , Leukotriene B4/physiology , Analysis of Variance , Animals , Disease Models, Animal , Macrophages , Mast Cells , Peritoneal Cavity/cytology , Rats , Sodium Chloride/pharmacology
11.
Int Arch Allergy Immunol ; 111(3): 244-52, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8917119

ABSTRACT

Recently we demonstrated that the eosinophil migration into rat peritoneal cavities induced by a large volume of saline is mediated by LTB4 released by the resident macrophages and mast cells. In the present study, we have investigated the involvement of IL-5 and IL-8 in this process. We observed that saline-stimulated mast cells released the eosinophil chemotactic cytokines IL-5 and IL-8, while macrophages released only IL-8. These observations were confirmed by the ability of antibodies against IL-5 and IL-8 block the eosinophil chemotactic activity of the mast cell supernatants while the chemotactic activity of the macrophage supernatants was inhibited only by the antibody to IL-8. Recombinant forms of IL-5 and IL-8, when injected intraperitoneally, induced a dose-dependent eosinophil accumulation in naïve rats. The mechanism by which these cytokines induce eosinophil migration seems to be dependent on the resident cell population since depleting the peritoneal cavities of the latter renders the animals unresponsive to the eosinophil recruitment when challenged with IL-5, IL-8 or the supernatants of saline-treated mast cells or macrophages. Dexamethasone and MK 886 blocked the eosinophil migration induced by both the supernatants of saline-stimulated mast cells or macrophages and by IL-5 or IL-8. The IL-5-induced eosinophil migration was also blocked by BW A4C, another lipoxygenase inhibitor. Together, these results suggest LTB4 to be the lipoxygenase metabolite involved in the eosinophil recruitment induced by IL-5 and IL-8. Our results indicate that the eosinophil migration induced by saline is a complex phenomenon which is dependent on the resident mast cells and macrophages and is mediated by LTB4, IL-5 and IL-8. Mast cells release LTB4, IL-5 and IL-8, whereas macrophages release mainly LTB4 and IL-8. The inhibition of one of these mediators (IL-5, IL-8 or LTB4) completely blocked the eosinophil migration induced by saline, suggesting that they act synergistically.


Subject(s)
Benzeneacetamides , Chemotaxis, Leukocyte/physiology , Eosinophils/physiology , Interleukin-5/physiology , Interleukin-8/physiology , Sodium Chloride/pharmacology , Animals , Cell Movement/drug effects , Chemotaxis, Leukocyte/drug effects , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Glucocorticoids/pharmacology , Hydroxamic Acids/pharmacology , Indoles/antagonists & inhibitors , Indoles/pharmacology , Interleukin-5/antagonists & inhibitors , Interleukin-5/pharmacology , Interleukin-8/pharmacology , Leukotriene B4/physiology , Lipoxygenase Inhibitors/pharmacology , Macrophages, Peritoneal/physiology , Male , Mast Cells/physiology , Peritoneal Cavity/cytology , Rats , Rats, Wistar , Recombinant Proteins
12.
Actual. pediátr ; 6(1): 40-3, mar. 1996.
Article in Spanish | LILACS | ID: lil-190426

ABSTRACT

La presencia de bacterias a nivel de la mucosa de las vías urinarias determina una respuesta inflamatoria. El factor responsable de la invasión de células inflamatorias es la IL6 (interleuquina 6) y los componentes bacterianos inductores son las adhesinas y el LPS de la membrana bacteriana. La actividad inflamatoria no es sólo responsable de los síntomas agudos sino de la eliminación bacteriana. En los estudios experimentales, los animales no reactivos al LPS fueron incapaces de eliminar las bacterias y en embarazadas se ha demostrado que una respuesta menor de IL6 puede relacionarse con la mayor incidencia de pielonefritis (1-4). La respuesta inflamatoria también ha sido relacionada con la producción de IL8 (interleuquina 8). Estudios en humanos han mostrado correlación entre concentración de IL8 urinaria y los recuentos leucocitarios (5).


Subject(s)
Humans , Pyelonephritis/diagnosis , Pyelonephritis/drug therapy , Pyelonephritis/etiology , Pyelonephritis/history , Pyelonephritis/immunology , Pyelonephritis/nursing , Pyelonephritis/physiopathology , Interleukin-6/immunology , Interleukin-6/isolation & purification , Interleukin-6/physiology , Interleukin-8/history , Interleukin-8/immunology , Interleukin-8/isolation & purification , Interleukin-8/physiology
14.
Br J Pharmacol ; 104(3): 765-7, 1991 Nov.
Article in English | MEDLINE | ID: mdl-1797337

ABSTRACT

1. The hyperalgesic effects of interleukin-8 (IL-8), interleukin-1 beta (IL-1 beta) and carrageenin were measured in a rat paw pressure test. 2. IL-8 evoked a dose-dependent hyperalgesia which was attenuated by a specific antiserum, the beta-adrenoceptor antagonists atenolol and propranolol, the dopamine receptor antagonist SCH 23390 and the adrenergic neurone-blocking agent guanethidine. The hyperalgesia was not attenuated by the cyclooxygenase inhibitor indomethacin or the IL-1 beta analogue Lys-D-Pro-Thr. 3. IL-1 beta-evoked hyperalgesia was attenuated by indomethacin and Lys-D-Pro-Thr but not by atenolol or SCH 23390. 4. Carrageenin-evoked hyperalgesia was attenuated by atenolol, indomethacin and anti-IL-8 serum. The effects of atenolol and anti-IL-8 serum were not additive. The effects of indomethacin and anti-IL-8 serum were additive: this combination abolished carrageenin-evoked hyperalgesia. 5. A new biological activity of IL-8 is described, namely the capacity to evoke hyperalgesia by a prostaglandin-independent mechanism. IL-8 is the first endogenous mediator to be identified as evoking hyperalgesia involving the sympathetic nervous system. Since IL-8 is released by activated macrophages and endothelial cells it may be a humoral link between tissue injury and sympathetic hyperalgesia.


Subject(s)
Interleukin-8/physiology , Pain/physiopathology , Sympathetic Nervous System/physiopathology , Animals , Atenolol/pharmacology , Benzazepines/pharmacology , Carrageenan , Dinoprostone/metabolism , Edema/chemically induced , Edema/complications , Edema/pathology , Foot/pathology , Guanethidine/pharmacology , Interleukin-1/pharmacology , Interleukin-8/antagonists & inhibitors , Interleukin-8/pharmacology , Male , Monocytes/drug effects , Monocytes/metabolism , Nociceptors/drug effects , Pain/etiology , Propranolol/pharmacology , Rats , Rats, Inbred Strains , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL