Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Pediatr Endocrinol Metab ; 31(6): 649-654, 2018 Jun 27.
Article in English | MEDLINE | ID: mdl-29723156

ABSTRACT

BACKGROUND: The receptor for advanced glycation end products (RAGE) has a critical role in the pathogenesis of inflammation. In healthy children, its basal expression on the peripheral blood mononuclear cell (PBMC) and the basal circulating soluble RAGE (sRAGE) levels are unknown. The aim of this study was to describe both. METHODS: This is a monocentric, observational and descriptive study of samples obtained from healthy children. The RAGE expression on PBMC was analyzed using flow cytometry. The sRAGE values were determined with a specific sandwich enzyme-linked immunosorbent assay (ELISA) kit, later the relation between cellular RAGE and sRAGE was described. RESULTS: Forty-three children were included. The median sRAGE level was 849.0±579.0 pg/mL. The RAGE mean fluorescence intensity (MFI) was 1382±506 in monocytes and 792±506 in lymphocytes. There were no differences between genders. A negative correlation was found between sRAGE and RAGE MFI in lymphocytes (r=-0.3; p=0.04). CONCLUSIONS: We describe for the first time the RAGE surface levels on PBMC in children. It showed a negative correlation with sRAGE. The sRAGE circulating level is lower than the sRAGE level described in adult population or non-healthy children. Our findings should be confirmed in order to apply them as reference values for future investigations.


Subject(s)
Leukocytes, Mononuclear/metabolism , Receptor for Advanced Glycation End Products/blood , Receptor for Advanced Glycation End Products/metabolism , Adolescent , Age Factors , Aging/blood , Aging/metabolism , Antigens, Surface/metabolism , Child , Child, Preschool , Female , Flow Cytometry , Healthy Volunteers , Humans , Male , Protein Isoforms/blood , Protein Isoforms/metabolism , Solubility
2.
Lung ; 193(1): 19-23, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25355250

ABSTRACT

PURPOSE: Traditional inflammatory biomarkers are insufficient for the evaluation of bronchiolitis severity. Recent investigations have shown that the receptor for advanced glycation end product (RAGE) and its soluble isoforms (sRAGE) play a critical role in the pathogenesis of lung injury. Main objective was to assess the serum levels of sRAGE of children with severe bronchiolitis admitted to the pediatric intensive care unit (PICU). Secondary objective was to study sRAGE correlation with the evolution and traditional biomarkers. METHODS: Prospective, observational and descriptive study, 43 healthy controls and 37 patients (December 2011-February 2012) were enrolled. sRAGE levels were assessed and compared. In patients, the relation between sRAGE levels and clinical evolution, respiratory assistance, white blood cell count, absolute neutrophils count, serum C-reactive protein, and serum procalcitonin was analyzed. RESULTS: A statistical difference was found in the mean value of sRAGE at PICU admission between patients and controls (1,215.7 ± 535 vs 849 ± 579 pg/ml). Also a significant inverse correlation was found between sRAGE and the Wood-Downes Score at admission (p = 0.02). CONCLUSIONS: Serum sRAGE could be elevated in children with bronchiolitis. Larger clinical studies are necessary to elucidate its role as a bronchiolitis inflammatory and/or lung injury biomarker.


Subject(s)
Bronchiolitis/blood , Receptors, Immunologic/blood , Age Factors , Biomarkers/blood , Bronchiolitis/diagnosis , Case-Control Studies , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Intensive Care Units, Pediatric , Length of Stay , Male , Patient Admission , Pilot Projects , Predictive Value of Tests , Prognosis , Prospective Studies , Receptor for Advanced Glycation End Products , Severity of Illness Index , Time Factors , Up-Regulation
3.
J Immunol ; 193(8): 4195-202, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25225662

ABSTRACT

Endotoxin tolerance (ET) is a state of reduced responsiveness to endotoxin stimulation after a primary bacterial insult. This phenomenon has been described in several pathologies, including sepsis, in which an endotoxin challenge results in reduced cytokine production. In this study, we show that the NFκ L chain enhancer of activated B cells 2 (NFκB2)/p100 was overexpressed and accumulated in a well-established in vitro human monocyte model of ET. The p100 accumulation in these cells inversely correlated with the inflammatory response after LPS stimulation. Knocking down NFκB2/p100 using small interfering RNA in human monocytes further indicated that p100 expression is a crucial factor in the progression of ET. The monocytes derived from patients with sepsis had high levels of p100, and a downregulation of NFκB2/p100 in these septic monocytes reversed their ET status.


Subject(s)
Endotoxins/immunology , Immune Tolerance , Monocytes/immunology , NF-kappa B p52 Subunit/biosynthesis , Sepsis/immunology , Aged , Down-Regulation , Gene Knockout Techniques , Humans , Inflammation/immunology , NF-kappa B p52 Subunit/genetics , RNA Interference , RNA, Small Interfering
4.
Biochem Biophys Res Commun ; 423(2): 331-7, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22659741

ABSTRACT

Monocyte exposure to tumor cells induces a transient state in which these cells are refractory to further exposure to cancer. This phenomenon, termed "tumor tolerance", is characterized by a decreased production of proinflammatory cytokines in response to tumors. In the past, we found that this effect comprises IRAK-M up regulation and TLR4 and CD44 activation. Herein we have established a human model of tumor tolerance and have observed a marked down-regulation of MHCII molecules as well as the MHCII master regulator, CIITA, in monocytes/macrophages. These cells combine an impaired capability for antigen presentation with potent phagocytic activity and exhibit an M2-like phenotype. In addition circulating monocytes isolated from Chronic Lymphocytic Leukemia patients exhibited the same profile as tumor tolerant cells after tumor ex vivo exposition.


Subject(s)
Antigen Presentation , Immune Tolerance , Models, Biological , Monocytes/immunology , Neoplasms/immunology , Phagocytosis , HeLa Cells , Histocompatibility Antigens Class II/immunology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lipopolysaccharides/immunology , Macrophages/immunology , Nuclear Proteins/immunology , Trans-Activators/immunology
5.
J Immunol ; 188(8): 3584-93, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22427642

ABSTRACT

Blood monocytes recognize Gram-negative bacteria through the TLR4, which signal via MyD88- and TRIF-dependent pathway to trigger an immune-inflammatory response. However, a dysregulated inflammatory response by these cells often leads to severe pathologies such as sepsis. We investigated the role of CD16 in the regulation of human monocyte response to Gram-negative endotoxin and sepsis. Blood monocytes from sepsis patients demonstrated an upregulation of several TRIF-dependent genes as well as a selective expansion of CD16-expressing (CD16(+)) monocytes. Gene expression and biochemical studies revealed CD16 to regulate the TRIF-dependent TLR4 pathway in monocytes by activating Syk, IFN regulatory factor 3, and STAT1, which resulted in enhanced expression of IFNB, CCL5, and CXCL10. CD16 also upregulated the expression of IL-1R-associated kinase M and IL-1 receptor antagonist, which are negative regulators of the MyD88-dependent pathway. CD16 overexpression or small interfering RNA knockdown in monocytes confirmed the above findings. Interestingly, these results were mirrored in the CD16(+) monocyte subset isolated from sepsis patients, providing an in vivo confirmation to our findings. Collectively, the results from the current study demonstrate CD16 as a key regulator of the TRIF-dependent TLR4 pathway in human monocytes and their CD16-expressing subset, with implications in sepsis.


Subject(s)
Gene Expression Regulation/immunology , Monocytes/metabolism , Receptors, IgG/genetics , Sepsis/immunology , Adaptive Immunity , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Adult , Animals , Endotoxins/pharmacology , Gene Expression Regulation/drug effects , Humans , Interferons/genetics , Interferons/immunology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Mice , Mice, Inbred C57BL , Middle Aged , Monocytes/immunology , Monocytes/pathology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Primary Cell Culture , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/immunology , RNA, Small Interfering/genetics , Receptors, IgG/immunology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , Sepsis/genetics , Sepsis/pathology , Signal Transduction/drug effects , Signal Transduction/immunology , Syk Kinase , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Transfection
6.
J Leukoc Biol ; 91(6): 933-45, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22459945

ABSTRACT

The MMPs constitute a family of endopeptidases that can cleavage extracellular proteins. They are involved in a number of events; some of these include inflammatory processes. One of its targets is the TREM-1, which has emerged as an important modulator of innate immune responses in mammals. This transmembrane glycoprotein possesses an Ig-like ectodomain readily shed by MMPs to generate sTREM-1. Whereas membrane-anchored TREM-1 amplifies inflammatory responses, sTREM-1 exhibits anti-inflammatory properties. Here we show that sustained cell surface expression of TREM-1 in human monocytes, through metalloproteinase inhibition, counteracts the well-characterized down-regulation of several proinflammatory cytokines during the ET time-frame, also known as M2 or alternative activation. In addition to the cytokines profile, other features of the ET phenotype were underdeveloped when TREM-1 was stabilized at the cell surface. These events were mediated by the signal transducers PI3Ks and Syk. We also show that sTREM-1 counteracts the proinflammatory response obtained by membrane TREM-1 stabilization but failed to induce ET on naïve human monocytes. As the sustained TREM-1 expression at the cell surface suffices to block the progress of a refractory state in human monocytes, our data indicate that TREM-1 and MMPs orchestrate an "adaptive" form of innate immunity by modulating the human monocytes response to endotoxin.


Subject(s)
Collagenases/immunology , Membrane Glycoproteins/immunology , Monocytes/immunology , NF-kappa B/immunology , Phosphatidylinositol 3-Kinases/immunology , Receptors, Immunologic/immunology , Signal Transduction , Cytokines/immunology , Endotoxins/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Immunity, Innate/drug effects , Inflammation/immunology , Protein Stability , Triggering Receptor Expressed on Myeloid Cells-1
7.
PLoS One ; 6(12): e29577, 2011.
Article in English | MEDLINE | ID: mdl-22216320

ABSTRACT

Cystic Fibrosis (CF) is an inherited pleiotropic disease that results from abnormalities in the gene codes of a chloride channel. The lungs of CF patients are chronically infected by several pathogens but bacteraemia have rarely been reported in this pathology. Besides that, circulating monocytes in CF patients exhibit a patent Endotoxin Tolerance (ET) state since they show a significant reduction of the inflammatory response to bacterial stimulus. Despite a previous description of this phenomenon, the direct cause of ET in CF patients remains unknown. In this study we have researched the possible role of microbial/endotoxin translocation from a localized infection to the bloodstream as a potential cause of ET induction in CF patients. Plasma analysis of fourteen CF patients revealed high levels of LPS compared to healthy volunteers and patients who suffer from Chronic Obstructive Pulmonary Disease. Experiments in vitro showed that endotoxin concentrations found in plasma of CF patients were enough to induce an ET phenotype in monocytes from healthy controls. In agreement with clinical data, we failed to detect bacterial DNA in CF plasma. Our results suggest that soluble endotoxin present in bloodstream of CF patients causes endotoxin tolerance in their circulating monocytes.


Subject(s)
Cystic Fibrosis/blood , Endotoxins/pharmacology , Lipopolysaccharides/pharmacology , Monocytes/drug effects , Base Sequence , Biological Transport , Case-Control Studies , DNA Primers , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Humans , Lipopolysaccharides/metabolism
9.
J Immunol ; 182(10): 6494-507, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19414804

ABSTRACT

Monocyte exposure to LPS induces a transient state in which these cells are refractory to further endotoxin stimulation. This phenomenon, termed endotoxin tolerance (ET), is characterized by a decreased production of cytokines in response to the proinflammatory stimulus. We have established a robust model of ET and have determined the time frame and features of LPS unresponsiveness in cultured human monocytes. A large number of genes transcribed in tolerant monocytes were classified as either "tolerizable" or "nontolerizable" depending on their expression levels during the ET phase. Tolerant monocytes exhibit rapid IL-1R-associated kinase-M (IRAK-M) overexpression, high levels of triggering receptor expressed on myeloid cells-1 (TREM-1) and CD64, and a marked down-regulation of MHC molecules and NF-kappaB2. These cells combine potent phagocytic activity with impaired capability for Ag presentation. We also show that circulating monocytes isolated from cystic fibrosis patients share all the determinants that characterize cells locked in an ET state. These findings identify a new mechanism that contributes to impaired inflammation in cystic fibrosis patients despite a high frequency of infections. Our results indicate that a tolerant phenotype interferes with timing, efficiency, and outcome of the innate immune responses against bacterial infections.


Subject(s)
Antigen Presentation/immunology , Cystic Fibrosis/immunology , Immune Tolerance , Lipopolysaccharides/immunology , Monocytes/immunology , Phagocytosis/immunology , Adult , Cystic Fibrosis/physiopathology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Gene Expression , Humans , Interleukin-10/biosynthesis , Interleukin-10/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , Male , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , RNA, Messenger/analysis , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
10.
PLoS One ; 3(7): e2667, 2008 Jul 16.
Article in English | MEDLINE | ID: mdl-18628981

ABSTRACT

Cystic Fibrosis (CF) is an inherited pleiotropic disease that results from abnormalities in the gene that codes for the chloride channel, Cystic Fibrosis Transmembrane Conductance Regulator (CFTR). CF patients are frequently colonized by several pathogens, but the mechanisms that allow colonization in spite of apparently functional immune systems are incompletely understood. In this paper we show that blood peripheral monocytes isolated from CF patients are found in an endotoxin tolerance state, yet this is not due to a deficient TLR activation. On the other hand, levels of the amplifier of inflammatory responses, TREM-1 (Triggering Receptor Expressed on Myeloid cells), are notably down-regulated in monocytes from patients, in comparison to those extracted from healthy volunteers. Furthermore, the soluble form of TREM-1 (sTREM-1) was not detected in the sera of patients. Additionally, and in strict contrast to patients who suffer from Chronic Obstructive Pulmonary Disease (COPD), CF monocytes challenged ex vivo with LPS neither up-regulated membrane-anchored TREM-1 nor sTREM-1. Finally, similar levels of PGE(2) expression and p65 translocation into the nucleus were found in both patients and healthy volunteers, thus suggesting that TREM-1 regulation is neither controlled by PGE(2) levels nor by p65 activation in this case. However, PU.1 translocation into the nucleus was significantly higher in CF monocytes than in controls, suggesting a role for this transcription factor in the control of TREM-1 expression. We conclude that down-regulation of TREM-1 expression in cystic fibrosis patients is at least partly responsible for the endotoxin tolerance state in which their monocytes are locked.


Subject(s)
Cystic Fibrosis/blood , Down-Regulation , Lipopolysaccharides/metabolism , Membrane Glycoproteins/biosynthesis , Monocytes/cytology , Receptors, Immunologic/biosynthesis , Adult , Aged , Case-Control Studies , Cell Separation , Dinoprostone/metabolism , Endotoxins/metabolism , Female , Humans , Lung/microbiology , Male , Triggering Receptor Expressed on Myeloid Cells-1
11.
Med Oncol ; 24(4): 436-44, 2007.
Article in English | MEDLINE | ID: mdl-17917095

ABSTRACT

Although the complex and multifactorial process of tumour growth has been extensively studied for decades, our understanding of the fundamental relationship between tumour growth dynamics and genetic expression profile remains incomplete. Recent studies of tumour dynamics indicate that gene expression in solid tumours would depend on the distance from the centre of the tumour. Since tumour proliferative activity is mainly localised to its external zone, and taking into account that generation and expansion of genetic mutations depend on the number of cell divisions, important differences in gene expression between central and peripheral sections of the same tumour are to be expected. Here, we have studied variations in the genetic expression profile between peripheral and internal samples of the same brain tumour. We have carried out microarray analysis of mRNA expression, and found a differential profile of genetic expression between the two cell subsets. In particular, one major nuclear protein that regulates cell responses to DNA-damaging and stress signals, GADD45alpha, was expressed at much lower levels in the peripheral zone, as compared to tumour core samples. These differences in GADD45alpha mRNA transcription levels have been confirmed by quantitative analysis via real time PCR, and protein levels of GADD45alpha also exhibit the same pattern of differential expression. Our findings suggest that GADD45alpha might play a major role in the regulation of brain tumour invasive potential.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Cycle Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Brain Neoplasms/genetics , Cell Cycle Proteins/genetics , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Profiling , Magnetic Resonance Imaging , Neoplasm Invasiveness , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats , Rats, Wistar , Transcription, Genetic
12.
J Immunol ; 179(6): 4065-73, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17785845

ABSTRACT

Triggering receptors expressed on myeloid cell (TREM) proteins are a family of cell surface receptors that participate in diverse cellular processes such as inflammation, coagulation, and bone homeostasis. TREM-1, in particular, is expressed on neutrophils and monocytes and is a potent amplifier of inflammatory responses. LPS and other microbial products induce up-regulation of cell surface-localized TREM-1 and the release of its soluble form, sTREM-1. Two hypotheses have been advanced to explain the origin of sTREM-1: alternative splicing of TREM-1 mRNA and proteolytic cleavage(s) of mature, membrane-anchored TREM-1. In this report, we present conclusive evidence in favor of the proteolytic mechanism of sTREM-1 generation. No alternative splicing forms of TREM-1 were detected in monocytes/macrophages. Besides, metalloproteinase inhibitors increased the stability of TREM-1 at the cell surface while significantly reducing sTREM-1 release in cultures of LPS-challenged human monocytes and neutrophils. We conclude that metalloproteinases are responsible for shedding of the TREM-1 ectodomain through proteolytic cleavage of its long juxtamembrane linker.


Subject(s)
Lipopolysaccharides/immunology , Metalloproteases/metabolism , Monocytes/metabolism , Receptors, Immunologic/metabolism , Alternative Splicing/genetics , Base Sequence , Cells, Cultured , Humans , Hydrolysis , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/metabolism , Metalloproteases/chemistry , Molecular Sequence Data , Molecular Weight , Monocytes/enzymology , Monocytes/immunology , Protease Inhibitors/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary/genetics , RNA, Messenger/isolation & purification , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/genetics , Salmonella/immunology , Solubility
13.
J Endotoxin Res ; 13(1): 39-52, 2007.
Article in English | MEDLINE | ID: mdl-17621545

ABSTRACT

Acute coronary syndrome (ACS) groups different cardiac diseases whose development is associated with inflammation. Here we have analyzed the levels of inflammatory cytokines and of members of the TLR/IRAK pathway including IRAK-M in monocytes from ACS patients classified as either UA (unstable angina), STEMI (ST-elevation myocardial infarction) or NSTEMI (non-ST-elevation myocardial infarction). Circulating monocytes from all patients, but not from healthy individuals, showed high levels of pro-inflammatory cytokines, TNF-alpha and IL-6, as well as of IRAK-M and IL-10. TLR4 was also up-regulated, but IRAK-1, IRAK-4 and MyD88 levels were similar in patients and controls. Further, we investigated the consequences of cytokines/IRAK-M expression on the innate immune response to endotoxin. Ex vivo responses to LPS were markedly attenuated in patient monocytes compared to controls. Control monocytes cultured for 6 h in supplemented medium (10% serum from ACS patients) expressed IRAK-M, and LPS stimulation failed to induce TNF-alpha and IL-6 in these cultures. Pre-incubation of the serum with a blocking anti-TNF-alpha antibody reduced this endotoxin tolerance effect, suggesting that TNF-alpha controls this phenomenon, at least partially. We show for the first time that inflammatory responses associated with ACS induce an unresponsiveness state to endotoxin challenge in circulating monocytes, which correlates with expression of IRAK-M, TLR4 and IL-10. The magnitude of this response varies according to the clinical condition (UA, STEMI or NSTEMI), and is regulated by TNF-alpha.


Subject(s)
Angina, Unstable/immunology , Gene Expression Regulation/immunology , Interleukin-1 Receptor-Associated Kinases/immunology , Lipopolysaccharides/immunology , Monocytes/immunology , Myocardial Infarction/immunology , Angina, Unstable/metabolism , Angina, Unstable/pathology , Antibodies, Blocking/pharmacology , Drug Tolerance/immunology , Female , Humans , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Male , Middle Aged , Monocytes/drug effects , Monocytes/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/immunology
14.
J Leukoc Biol ; 82(3): 700-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17576820

ABSTRACT

Osteoclasts are large, multinucleated cells, which originate from the fusion of macrophages. They play a central role in bone development and remodeling via the resorption of bone and are thus important mediators of bone loss, which leads to osteoporosis. IL-1R-associated kinase (IRAK)-M is a pseudokinase, which acts as a negative modulator of innate immune responses mediated by TLRs and IL-1R. Recently, it has been reported that IRAK-M also participates in the control of macrophage differentiation into osteoclasts. In addition, it was shown that IRAK-M knockout mice develop a strong osteoporosis phenotype, suggesting that down-regulation of this molecule activates osteoclast-mediated bone resorption. We studied the effect of the osteoporosis-inducing glucocorticoid, 6-methylprednisolone (6-MP), on IRAK-M expression in osteoclasts. Our results showed that osteoclasts, derived from THP-1 and RAW cells as well as human blood monocytes, differentiated into osteoclasts, express high levels of IRAK-M at mRNA and protein levels. In addition, 6-MP down-regulates IRAK-M expression, which correlates with an increased activation of bone resorption. These findings suggest a mechanism of corticosteroid-induced osteoporosis and open new avenues for treating this endemic disease of Western societies.


Subject(s)
Bone Resorption , Glucocorticoids/pharmacology , Interleukin-1 Receptor-Associated Kinases/metabolism , Methylprednisolone/pharmacology , Osteoclasts/drug effects , Osteoporosis/metabolism , Adrenal Cortex Hormones/pharmacology , Animals , Blotting, Western , Cell Differentiation , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Down-Regulation , Humans , Immunoenzyme Techniques , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Osteoporosis/chemically induced , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tartrates/pharmacology
15.
J Endotoxin Res ; 12(5): 296-306, 2006.
Article in English | MEDLINE | ID: mdl-17059693

ABSTRACT

In contrast to the thoroughly characterized mechanisms of positive regulation within cytokine signaling pathways, our knowledge of negative feedback loops is comparatively sparse. We and others have previously reported that IRAK-M down-regulates inflammatory responses to multiple stimuli. In particular, we could show that the nitric oxide (NO) donor, GSNO, induces IRAK-M overexpression in human monocytes. Here we study the expression of another important negative regulator of cytokine signaling, SOCS-1, in human monocytes exposed to GSNO. The NO donor induced significant levels of SOCS-1 mRNA and protein, 6 h and 16 h after stimulation, respectively. Monocytes stimulated with GSNO for longer periods (24 h and 48 h) failed to express IL-6 and IP-10 upon LPS challenge. In addition, and in line with previous reports of NO-mediated induction of TNF-alpha, we have found that exposure to this cytokine induces SOCS-1 mRNA in human monocytes. A blocking antibody against TNF-alpha impaired SOCS-1 expression upon GSNO treatment and re-instated IL-6 and IP-10 mRNA levels after LPS challenge in cultures pretreated with the NO donor. We conclude that NO stimulates SOCS-1 overexpression in a pathway at least partially regulated by TNF-alpha.


Subject(s)
Monocytes/drug effects , Monocytes/metabolism , Nitric Oxide/pharmacology , Suppressor of Cytokine Signaling Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cells, Cultured , Humans , Kinetics , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...