Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
2.
Free Radic Biol Med ; 172: 550-561, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34245858

ABSTRACT

The small GTPase Arf6 regulates many cellular processes, including cytoskeletal remodeling, receptor endocytosis, and pathogen phagocytosis. Arf6 silencing in neutrophil (PMN)-like cells is well-known to inhibit chemotactic peptide-mediated activation of phospholipase D, the oxidative burst, and ß2 integrin-dependent adhesion. In conditional knockout (cKO) mice, the migration to inflammatory sites of Arf6-deficient PMNs was diminished and associated with reduced cell surface expression of ß2 integrins. In this study we assessed the impact of Arf6 depletion on the functions and gene expression profile of PMNs isolated from the mouse air pouch. Numerous genes involved in response to oxygen levels, erythrocyte and myeloid differentiation, macrophage chemotaxis, response to chemicals, apoptosis, RNA destabilization, endosome organization, and vesicle transport were differentially expressed in PMNs cKO for Arf6. Lpar6 and Lacc-1 were the most up-regulated and down-regulated genes, respectively. The deletion of Arf6 also decreased Lacc-1 protein level in PMNs, and silencing of Arf6 in THP-1 monocytic cells delayed LPS-mediated Lacc-1 expression. We report that fMLP or zymosan-induced glycolysis and oxygen consumption rate were both decreased in air pouch PMNs but not in bone marrow PMNs of Arf6 cKO mice. Reduced oxygen consumption correlated with a decrease in superoxide and ROS production. Deletion of Arf6 in PMNs also reduced phagocytosis and interfered with apoptosis. The data suggest that Arf6 regulates energy metabolism, which may contribute to impaired phagocytosis, ROS production, and apoptosis in PMN-Arf6 cKO. This study provides new information on the functions and the inflammatory pathways influenced by Arf6 in PMNs.


Subject(s)
Neutrophils , Respiratory Burst , Animals , Energy Metabolism/genetics , Mice , Phagocytosis , Superoxides
3.
Sci Rep ; 11(1): 11248, 2021 05 27.
Article in English | MEDLINE | ID: mdl-34045571

ABSTRACT

The myeloid inhibitory receptor CLEC12A negatively regulates inflammation. Reduced CLEC12A expression enhances inflammation in CLEC12A knock-out mice with collagen antibody-induced arthritis. Moreover, CLEC12A internalisation augments human neutrophil activation. We thus postulated that CLEC12A expression on circulating myeloid cells of rheumatoid arthritis patients is associated with disease manifestations. Cell-surface, CLEC12A receptor expression was determined on circulating neutrophils and monocytes of eRA patients and of healthy donors. Generalized estimating equations model, Student's t-test and Spearman's correlations were performed to compare CLEC12A expression between groups and test its association with disease activity and clinical parameters. Plasma cytokines were measured by multiplex immunoassay. Patients with reduced neutrophil or monocyte CLEC12A expression at baseline and at 3 months have an increased simple disease activity index. Low baseline CLEC12A expression also correlates with a higher SDAI at 6 months. In contrast, positive correlations were observed between baseline CLEC12A expression and several cytokines. Moreover, neutrophil and monocyte CLEC12A expression is significantly higher in early rheumatoid arthritis patients at baseline than healthy controls. Circulating neutrophil and monocyte CLEC12A expression correlates with disease activity at baseline and is predictive of SDAI at later stages of the disease indicative of a regulatory role for CLEC12A in RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Cytokines/blood , Lectins, C-Type/metabolism , Myeloid Cells/metabolism , Receptors, Mitogen/metabolism , Aged , Arthritis, Rheumatoid/diagnosis , Female , Humans , Male , Middle Aged , Monocytes/metabolism , Neutrophil Activation , Neutrophils/metabolism , Severity of Illness Index
4.
J Immunol ; 206(3): 505-514, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33361205

ABSTRACT

High concentrations of the damage-associated molecular patterns S100A8 and S100A9 are found in skin and serum from patients suffering from psoriasis, an IL-17-related disease. Notably, although the expression of these proteins correlates with psoriatic disease severity, the exact function of S100A8 and S100A9 in psoriasis pathogenesis remains unclear. In this study, we investigated the role of S100A8 and S100A9 in psoriasis-associated skin hyperplasia and immune responses using S100a8-/- and S100a9-/- mice in an imiquimod-induced model of psoriasis. We found that S100a8-/- and S100a9-/- psoriatic mice exhibit worsened clinical symptoms relative to wild-type mice and increased expression of S100A9 and S100A8 proteins in keratinocytes, respectively. In addition, the loss of S100A8 enhances proliferation of keratinocytes and disrupts keratinocyte differentiation. We further detected elevated production of IL-17A and -F from CD4+ T cells in the absence of S100A8 and S100A9, as well as increased infiltration of neutrophils in the skin. In addition, treatment with anti-IL-17A and -F was found to reduce psoriasis symptoms and skin hyperplasia in S100a8-/- and S100a9-/- mice. These data suggest that S100A8 and S100A9 regulate psoriasis by inhibiting production of IL-17A and -F, thereby, to our knowledge, providing new insights into their biological functions.


Subject(s)
Calgranulin A/metabolism , Calgranulin B/metabolism , Interleukin-17/metabolism , Psoriasis/metabolism , Psoriasis/pathology , Skin/pathology , Th17 Cells/immunology , Animals , Antibodies, Blocking/metabolism , Calgranulin A/genetics , Calgranulin B/genetics , Cells, Cultured , Disease Models, Animal , Humans , Hyperplasia , Imiquimod , Interleukin-17/immunology , Mice, Inbred C57BL , Mice, Knockout , Psoriasis/chemically induced
5.
PLoS One ; 14(8): e0221528, 2019.
Article in English | MEDLINE | ID: mdl-31437241

ABSTRACT

Expressed strongly by myeloid cells, damage-associated molecular pattern (DAMP) proteins S100A8 and S100A9 are found in the serum of patients with infectious and autoimmune diseases. Compared to S100A9, the role of S100A8 is controversial. We investigated its biological activity in collagen-induced arthritis using the first known viable and fertile S100a8-deficient (S100a8-/-) mouse. Although comparable to the wild type (WT) in terms of lymphocyte distribution in blood and in the primary and secondary lymphoid organs, S100a8-/- mice had increased numbers of neutrophils, monocytes and dendritic cells in the blood and bone marrow, and these all expressed myeloid markers such as CD11b, Ly6G and CD86 more strongly. Granulocyte-macrophage common precursors were increased in S100a8-/- bone marrow and yielded greater numbers of macrophages and dendritic cells in culture. The animals also developed more severe arthritic disease leading to aggravated osteoclast activity and bone destruction. These findings were correlated with increased inflammatory cell infiltration and cytokine secretion in the paws. This study suggests that S100A8 is an anti-inflammatory DAMP that regulates myeloid cell differentiation, thereby mitigating the development of experimental arthritis.


Subject(s)
Arthritis, Experimental/pathology , Calgranulin A/deficiency , Myelopoiesis , Animals , Arthritis, Experimental/diagnostic imaging , Bone Marrow/pathology , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Calgranulin A/metabolism , Cartilage/pathology , Cell Differentiation , Dendritic Cells/metabolism , Female , Gene Deletion , Mice , Myeloid Cells/pathology
6.
Clin Transl Med ; 8(1): 19, 2019 Jun 05.
Article in English | MEDLINE | ID: mdl-31165299

ABSTRACT

BACKGROUND: Biological agents have allowed remarkable improvement in controlling autoimmune arthropathies, although none of the numerous biologics readily available represent a universal treatment standard. Moreover, classical and genetic predictors are currently unsatisfactory to predict individual response to a biologic, and the best treatment selection is still based on a trial-and-error approach. Here, we report a clinical case demonstrating the usefulness of examining the leukocytes' secretome of patients. We set up and standardized a protocol that examines a patient's immune responses to establish the secretome of the blood mononuclear leukocytes and personalize the biotherapy. CASE PRESENTATION: A 24-year-old woman was diagnosed with active early rheumatoid arthritis. The initial treatment regimen (prednisone, methotrexate, hydroxychloroquine, naproxen) was inefficient, as well as the anti-TNF adalimumab. The diagnosis was revised as possible rheumatoid arthritis-like psoriatic arthritis and adalimumab was replaced by abatacept (IgG1 Fc-CTLA-4) to no avail. Five years later, abatacept was replaced by the anti-IL-12/IL-23 ustekinumab with no objective control over the symptoms. The patient was thus enrolled in a prospective study based on the quantification of cytokines secreted by peripheral blood leukocytes stimulated with well-known immune activators of pattern recognition receptors and cytokine signalling. The results of this study revealed that plasma concentrations of cytokines were similar between the patient and healthy donors. In comparison to leukocytes from healthy donors, the patient's secretome showed a unique overproduction of IL-6. The anti-IL-6 receptor tocilizumab was, therefore, administered with a rapid improvement of her active psoriatic arthritis that remained dependent on low prednisone dosage. Clinical parameters progressively returned to normal levels and her quality of life was greatly improved, despite the major delay to begin the present personalized treatment. CONCLUSIONS: An efficient way to effectively treat patients with complex autoimmune arthropathies, and avoid irreversible disability, is to know their leukocytes' secretome to identify abnormally secreted cytokines and personalize their biotherapy, as exemplified by this case report.

7.
Scand J Trauma Resusc Emerg Med ; 25(1): 114, 2017 Nov 25.
Article in English | MEDLINE | ID: mdl-29178941

ABSTRACT

BACKGROUND: Following tissue injury after trauma, the activation of innate immune pathways results in systemic inflammation, organ failure and an increased risk of infections. The objective of this study was to characterize the kinetics of the S100A8/S100A9 complex, a new-recognized alarmin, as well as its soluble receptor sRAGE, over time after trauma as potential early biomarkers of the risk of organ damage. METHODS: We collected comprehensive data from consenting patients admitted to an ICU following severe trauma. The blood samples were taken at Day 0 (admission), Day1, 3 and 5 S100A8/A9 and sRAGE were measured by ELISA. Biomarkers levels were reported as median (IQR). RESULTS: Thirty-eight patients sustaining in majority a blunt trauma (89%) with a median ISS of 39 were included. In this cohort, the S100A8/A9 complex increased significantly over time (p = 0.001), but its levels increment over time (D0 to D5) was significantly smaller in patients developing infection (7.6 vs 40.1 mcg/mL, p = 0.011). The circulating level of sRAGE circulating levels decreased over time (p < 0.0001) and was higher in patients who remained in shock on day 3 (550 vs 918 pg/mL; p = 0.02) or 5 (498 vs 644 pg/mL; p = 0.045). Admission sRAGE levels were significantly higher in non-survivors (1694 vs 745 pg/mL; p = 0.015) and was higher in patients developing renal failure (1143 vs 696 pg/mL, p = 0.011). DISCUSSION: Our findings reveal an interesting association between the biomarker S100A8/9 least increase over time and the presence of infectious complication after trauma. We describe that the sRAGE decline over time is in relation with shock and markers of ischemic injury. We also confirm the association of sRAGE levels measured at admission with mortality and the development of renal failure. CONCLUSIONS: This work illustrates the importance of following the circulating level of biomarker overtime. The utilization of S1008/9 as a tool to stratify infection risk and trigger early interventions need to be validated prospectively.


Subject(s)
Calgranulin A/blood , Calgranulin B/blood , Multiple Organ Failure/blood , Multiple Organ Failure/etiology , Multiple Trauma/blood , Receptor for Advanced Glycation End Products/blood , Adult , Biomarkers/blood , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Inflammation/blood , Male , Middle Aged , Multiple Trauma/complications , Multiple Trauma/mortality , Risk , Time Factors , Young Adult
8.
Oncotarget ; 7(29): 44975-44990, 2016 Jul 19.
Article in English | MEDLINE | ID: mdl-27391444

ABSTRACT

Effector T cell migration through the tissue extracellular matrix (ECM) is an important step of the adaptive immune response and in the development of inflammatory diseases. However, the mechanisms involved in this process are still poorly understood. In this study, we addressed the role of a collagen receptor, the discoidin domain receptor 1 (DDR1), in the migration of Th17 cells. We showed that the vast majority of human Th17 cells express DDR1 and that silencing DDR1 or using the blocking recombinant receptor DDR1:Fc significantly reduced their motility and invasion in three-dimensional (3D) collagen. DDR1 promoted Th17 migration by activating RhoA/ROCK and MAPK/ERK signaling pathways. Interestingly, the RhoA/ROCK signaling module was required for MAPK/ERK activation. Finally, we showed that DDR1 is important for the recruitment of Th17 cells into the mouse dorsal air pouch containing the chemoattractant CCL20. Collectively, our results indicate that DDR1, via the activation of RhoA/ROCK/MAPK/ERK signaling axis, is a key pathway of effector T cell migration through collagen of perivascular tissues. As such, DDR1 can contribute to the development of Th17-dependent inflammatory diseases.


Subject(s)
Cell Movement/physiology , Discoidin Domain Receptor 1/metabolism , MAP Kinase Signaling System/physiology , Th17 Cells/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Female , Humans , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
9.
J Immunol ; 195(9): 4198-209, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26408663

ABSTRACT

Th17 cells are critical effectors in inflammation and tissue damage such as bone erosion, but the mechanisms regulating their activation in this process are not fully understood. In this study, we considered the cooperation between cytokine receptors and integrin pathways in Th17-osteoclast function. We found that human Th17 cells coexpress IL-7R and the collagen-binding integrin α2ß1 (CD49b), and IL-7 increases their adhesion to collagen via α2ß1 integrin. In addition, coengagement of the two receptors in human Th17 cells cooperatively enhanced their IL-17 production and their osteoclastogenic function. The functional cooperation between IL-7R and α2ß1 integrin involves activation of the JAK/PI3K/AKT (protein kinase B) and MAPK/ERK pathways. We also showed that IL-7-induced bone loss in vivo is associated with Th17 cell expansion. Moreover, blockade of α2ß1 integrin with a neutralizing mAb inhibited IL-7-induced bone loss and osteoclast numbers by reducing Th17 cell numbers in the bone marrow and reducing the production of IL-17 and the receptor activator of NF-κB ligand. Thus, the cooperation between IL-7R and α2ß1 integrin can represent an important pathogenic pathway in Th17-osteoclast function associated with inflammatory diseases.


Subject(s)
Bone Resorption/etiology , Integrin alpha2beta1/physiology , Receptors, Interleukin-7/physiology , Th17 Cells/physiology , Cell Adhesion , Cell Polarity , Collagen/pharmacology , Humans , Lymphocyte Activation , MAP Kinase Signaling System , Osteoclasts/physiology , Osteogenesis , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology
10.
J Immunol ; 195(9): 4426-37, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26385519

ABSTRACT

Recognition of viral dsRNA by endosomal TLR3 activates innate immune response during virus infection. Trafficking of TLR3 to the endolysosomal compartment arising from fusion of late endosome (LE) with lysosome is required for recognition and detection of pathogen associated molecular patterns, which results in activation of the TLR3-dependent signaling cascade. Existing knowledge about the mechanism(s) and cellular factor(s) governing TLR3 trafficking is limited. In the current study, we identified intracellular S100A9 protein as a critical regulator of TLR3 trafficking. S100A9 was required for maturation of TLR3 containing early endosome (EE) into LE, the compartment that fuses with lysosome to form the endolysosomal compartment. A drastic reduction in cytokine production was observed in S100A9-knockout (KO) primary macrophages following RNA virus infection and treatment of cells with polyinosinic-polycytidylic acid (polyIC; a dsRNA mimetic that acts as a TLR3 agonist). Mechanistic studies revealed colocalization and interaction of S100A9 with TLR3 following polyIC treatment. S100A9-TLR3 interaction was critical for maturation of TLR3 containing EE into LE because TLR3 could not be detected in the LE of polyIC-treated S100A9-KO macrophages. Subsequently, TLR3 failed to colocalize with its agonist (i.e., biotin-labeled polyIC) in S100A9-deficient macrophages. The in vivo physiological role of S100A9 was evident from loss of cytokine production in polyIC-treated S100A9-KO mice. Thus, we identified intracellular S100A9 as a regulator of TLR3 signaling and demonstrated that S100A9 functions during pre-TLR3 activation stages by facilitating maturation of TLR3 containing EE into LE.


Subject(s)
Calgranulin B/immunology , Macrophages/immunology , RNA Viruses/immunology , Toll-Like Receptor 3/immunology , Animals , Blotting, Western , Calgranulin B/genetics , Calgranulin B/metabolism , Cell Line , Cell Line, Tumor , Cells, Cultured , Female , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Interferon-beta/genetics , Interferon-beta/immunology , Interferon-beta/metabolism , Macrophages/metabolism , Macrophages/virology , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Poly I-C/immunology , Poly I-C/pharmacology , Protein Transport/drug effects , Protein Transport/immunology , RNA Interference , RNA Viruses/physiology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 3/metabolism
11.
J Immunol Res ; 2015: 296149, 2015.
Article in English | MEDLINE | ID: mdl-27057553

ABSTRACT

S100A8/A9 (calprotectin) and S100A12 proinflammatory mediators are found at inflammatory sites and in the serum of patients with inflammatory or autoimmune diseases. These cytoplasmic proteins are secreted by neutrophils at sites of inflammation via alternative secretion pathways of which little is known. This study examined the nature of the stimuli leading to S100A8/A9 and S100A12 secretion as well as the mechanism involved in this alternative secretion pathway. Chemotactic agents, cytokines, and particulate molecules were used to stimulate human neutrophils. MSU crystals, PMA, and H2O2 induced the release of S100A8, S100A9, and S100A12 homodimers, as well as S100A8/A9 heterodimer. High concentrations of S100A8/A9 and S100A12 were secreted in response to nanoparticles like MSU, silica, TiO2, fullerene, and single-wall carbon nanotubes as well as in response to microbe-derived molecules, such as zymosan or HKCA. However, neutrophils exposed to the chemotactic factors fMLP failed to secrete S100A8/A9 or S100A12. Secretion of S100A8/A9 was dependent on the production of reactive oxygen species and required K(+) exchanges through the ATP-sensitive K(+) channel. Altogether, these findings suggest that S100A12 and S100A8/A9 are secreted independently either via distinct mechanisms of secretion or following the activation of different signal transduction pathways.


Subject(s)
Calgranulin A/metabolism , Calgranulin B/metabolism , Neutrophils/metabolism , Potassium/metabolism , Reactive Oxygen Species/metabolism , S100A12 Protein/metabolism , Antioxidants/pharmacology , Blotting, Western , Calgranulin A/chemistry , Calgranulin B/chemistry , Cytokines/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Ion Transport/drug effects , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Oxidants/pharmacology , Protein Multimerization/drug effects , S100A12 Protein/chemistry , Signal Transduction/drug effects , Silicon Dioxide/pharmacology , Titanium/pharmacology , Uric Acid/pharmacology
12.
J Immunol ; 191(12): 5941-50, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24244022

ABSTRACT

Th17 cells play a critical role in the pathogenesis of rheumatoid arthritis (RA), but the mechanisms by which these cells regulate the development of RA are not fully understood. We have recently shown that α2ß1 integrin, the receptor of type I collagen, is the major collagen-binding integrin expressed by human Th17 cells. In this study, we examined the role of α2ß1 integrin in Th17-mediated destructive arthritis in the murine model of collagen-induced arthritis (CIA). We found that α2ß1 integrin is expressed on synovial Th17 cells from CIA mice and its neutralization with a specific mAb significantly reduced inflammation and cartilage degradation, and protected the mice from bone erosion. Blockade of α2ß1 integrin led to a decrease in the number of Th17 cells in the joints and to a reduction of IL-17 levels in CIA mice. This was associated with an inhibition of receptor activator of NF-κB ligand levels and osteoclast numbers, and reduction of bone loss. We further show that α2ß1 integrin is expressed on synovial Th17 cells from RA patients, and that its ligation with collagen costimulated the production of IL-17 by polarized human Th17 cells by enhancing the expression of retinoic acid receptor-related orphan receptor C through ERK and PI3K/AKT. Our findings provide the first evidence, to our knowledge, that α2ß1 integrin is an important pathway in Th17 cell activation in the pathogenesis of CIA, suggesting that its blockade can be beneficial for the treatment of RA and other Th17-associated autoimmune diseases.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Arthritis, Experimental/therapy , Arthritis, Rheumatoid/metabolism , Integrin alpha2beta1/physiology , Osteolysis/prevention & control , Receptors, Collagen/physiology , Th17 Cells/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibody Specificity , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/immunology , Cartilage, Articular/pathology , Collagen/pharmacology , Cricetinae , Down-Regulation , Female , Humans , Inflammation , Integrin alpha2beta1/antagonists & inhibitors , Interleukin-17/blood , Lymphocyte Activation , MAP Kinase Signaling System , Mice , Mice, Inbred DBA , NF-kappa B/physiology , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Osteoclasts/pathology , Osteolysis/etiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , RANK Ligand/blood , Receptors, Collagen/antagonists & inhibitors , Signal Transduction , Synovial Membrane/metabolism , Synovial Membrane/pathology , Th17 Cells/physiology
13.
PLoS One ; 7(9): e45478, 2012.
Article in English | MEDLINE | ID: mdl-23029038

ABSTRACT

OBJECTIVE: The S100A9 and S100A8 proteins are highly expressed by neutrophils and monocytes and are part of a group of damage-associated molecular pattern molecules that trigger inflammatory responses. Sera and synovial fluids of patients with rheumatoid arthritis (RA) contain high concentrations of S100A8/A9 that correlate with disease activity. METHODS: In this study, we investigated the importance of S100A9 in RA by using neutralizing antibodies in a murine lipopolysaccharide-synchronized collagen-induced arthritis model. We also used an in vitro model of stimulation of human immune cells to decipher the role played by S100A9 in leukocyte migration and pro-inflammatory cytokine secretion. RESULTS: Treatment with anti-S100A9 antibodies improved the clinical score by 50%, diminished immune cell infiltration, reduced inflammatory cytokines, both in serum and in the joints, and preserved bone/collagen integrity. Stimulation of neutrophils with S100A9 protein led to the enhancement of neutrophil transendothelial migration. S100A9 protein also induced the secretion by monocytes of proinflammatory cytokines like TNFα, IL-1ß and IL-6, and of chemokines like MIP-1α and MCP-1. CONCLUSION: The effects of anti-S100A9 treatment are likely direct consequences of inhibiting the S100A9-mediated promotion of neutrophil transmigration and secretion of pro-inflammatory cytokines from monocytes. Collectively, our results show that treatment with anti-S100A9 may inhibit amplification of the immune response and help preserve tissue integrity. Therefore, S100A9 is a promising potential therapeutic target for inflammatory diseases like rheumatoid arthritis for which alternative therapeutic strategies are needed.


Subject(s)
Arthritis, Experimental/immunology , Calgranulin B/immunology , Inflammation/immunology , Leukocyte L1 Antigen Complex/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Arthritis, Experimental/metabolism , Autoantibodies/blood , Autoantibodies/immunology , Bone and Bones/pathology , Calgranulin B/metabolism , Cartilage/pathology , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cytokines/immunology , Cytokines/metabolism , Female , Humans , Inflammation/metabolism , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Leukocyte L1 Antigen Complex/metabolism , Mice , Monocytes/drug effects , Monocytes/immunology , Neutrophils/drug effects , Neutrophils/immunology , Transendothelial and Transepithelial Migration/drug effects , Transendothelial and Transepithelial Migration/immunology , Tumor Necrosis Factor-alpha/pharmacology
14.
J Allergy Clin Immunol ; 119(2): 391-7, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17291856

ABSTRACT

BACKGROUND: Up to 30% of asthmatic subjects are smokers, and smoking might be an important contributor to asthma pathology. Inducible nitric oxide synthase (iNOS), ornithine decarboxylase (ODC), and arginase I are involved in the arginine pathway. We have shown that arginase I and iNOS are upregulated in asthma. Smoking asthmatic subjects are reported to have low exhaled nitric oxide levels. The effect of cigarette smoking on the expression of arginase I in asthma is unknown. OBJECTIVES: The aims of this study were to investigate the expression of arginase I, ODC, and iNOS in asthmatic airways of smokers and nonsmokers and in vitro after nicotine stimulation. METHODS: Endobronchial biopsies were performed on 24 steroid-naive subjects with mild asthma: 12 smokers and 12 nonsmokers. Arginase I, ODC, and iNOS levels were assessed by means of immunohistochemistry and in situ hybridization (arginase I). In vitro stimulation of airway cells with nicotine was performed, followed by real-time PCR. RESULTS: Arginase I, ODC, and iNOS were expressed in the epithelium and smooth muscle bundles of both subgroups of asthmatic subjects. There was an increase of arginase I and ODC immunoreactivities in smoking compared with nonsmoking asthmatic subjects. There was no significant difference in immunoreactivity for iNOS between groups. Nicotine induced a 2-fold increase in arginase I and ODC expression in airway epithelial cells and fibroblasts. CONCLUSION: This study demonstrates that the expression of arginase I and ODC is increased in airways of smoking compared with nonsmoking asthmatic subjects and in vitro by nicotine. CLINICAL IMPLICATIONS: Increased expression of arginase I might lead to low exhaled nitric oxide and chronic obstructive pulmonary disease-like airway remodeling in smoking asthmatic subjects.


Subject(s)
Arginase/genetics , Arginine/metabolism , Asthma/metabolism , Bronchi/metabolism , Smoking/metabolism , Adolescent , Adult , Female , Humans , Immunohistochemistry , Male , Nicotine/pharmacology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/analysis , Nitric Oxide Synthase Type II/genetics , Ornithine Decarboxylase/analysis , Ornithine Decarboxylase/genetics , RNA, Messenger/analysis
17.
Infect Immun ; 70(12): 7073-80, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12438388

ABSTRACT

Oral candidiasis is a collective name for a group of disorders caused by the dimorphic fungus Candida albicans. Host defenses against C. albicans essentially fall into two categories: specific immune mechanisms and local oral mucosal epithelial cell defenses. Since oral epithelial cells secrete a variety of cytokines and chemokines in response to oral microorganisms and since C. albicans is closely associated with oral epithelial cells as a commensal organism, we wanted to determine whether interleukin-18 (IL-18) and gamma interferon (IFN-gamma) were produced by oral epithelial cells in response to C. albicans infection and lipopolysaccharide (LPS) stimulation. Our results showed that IL-18 mRNA and protein were constitutively expressed by oral epithelial cells and were down-regulated by Candida infections but increased following LPS stimulation. Both C. albicans and LPS significantly decreased pro-IL-18 (24 kDa) levels and increased active IL-18 (18 kDa) levels. This effect was IL-1beta-converting-enzyme dependent. The increase in active IL-18 protein levels promoted the production of IFN-gamma by infected cells. No effect was obtained with LPS. Although produced only at an early stage, secreted IFN-gamma seemed to be a preferential response by oral epithelial cells to C. albicans growth. These results provide additional evidence for the contribution of oral epithelial cells to local (direct contact) and systemic (IL-18 and IFN-gamma production) defense against exogenous stimulation such as C. albicans infection or LPS stimulation.


Subject(s)
Candida albicans/pathogenicity , Epithelial Cells/immunology , Interferon-gamma/biosynthesis , Interleukin-18/biosynthesis , Lipopolysaccharides/pharmacology , Mouth Mucosa/immunology , Candida albicans/growth & development , Cell Adhesion , Cells, Cultured , Epithelial Cells/microbiology , Humans , Lipopolysaccharides/immunology , Mouth Mucosa/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...