Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Redox Biol ; 67: 102898, 2023 11.
Article in English | MEDLINE | ID: mdl-37757542

ABSTRACT

TNFα-mediated signaling pathways play a pivotal role in the pathogenesis of inflammatory diseases such as rheumatoid arthritis (RA) and inflammatory bowel disease (IBD) by promoting phagocyte inflammatory functions, notably cytokine release and reactive oxygen species (ROS) production by NOX2. In contrast, interleukin-10 (IL-10), a powerful anti-inflammatory cytokine, potently shuts down phagocyte activation, making IL-10 an attractive therapeutic candidate. However, IL-10 therapy has shown limited efficacy in patients with inflammatory diseases. Here, we report that TNFα blocks IL-10 anti-inflammatory pathways in human monocytes, thereby prolonging inflammation. TNFα decreased IL-10-induced phosphorylation of STAT3 and consequently IL-10-induced expression of the major anti-inflammatory factor, SOCS3. Decreased STAT3 phosphorylation was due to a SHP1/2 phosphatase, as NSC-87877, a SHP1/2 inhibitor, restored STAT3 phosphorylation and prevented the TNFα-induced inhibition of IL-10 signaling. TNFα activated only SHP1 in human monocytes and this activation was NOX2-dependent, as diphenyleneiodonium, a NOX2 inhibitor, suppressed SHP1 activation and STAT3 dephosphorylation triggered by TNFα. ROS-induced activation of SHP1 was mediated by the redox-sensitive kinase, Lyn, as its inhibition impeded TNFα-induced SHP1 activation and STAT3 dephosphorylation. Furthermore, H2O2 recapitulated TNFα-inhibitory activity on IL-10 signaling. Finally, NSC-87877 dampened collagen antibody-induced arthritis (CAIA) in mice. These results reveal that TNFα disrupts IL-10 signaling by inducing STAT3 dephosphorylation through a NOX2-ROS-Lyn-SHP1 axis in human monocytes and that inhibition of SHP1/2 in vivo protects against CAIA. These new findings might explain the poor efficacy of IL-10 therapy in patients with inflammatory diseases and suggest that anti-TNFα agents and SHP1/2 inhibitors could improve the therapeutic use of IL-10.


Subject(s)
Interleukin-10 , Monocytes , Humans , Animals , Mice , Monocytes/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Tumor Necrosis Factor-alpha/metabolism , Reactive Oxygen Species/metabolism , Hydrogen Peroxide/metabolism , Cytokines/metabolism , Anti-Inflammatory Agents , STAT3 Transcription Factor/metabolism
2.
Biomedicines ; 10(2)2022 Jan 29.
Article in English | MEDLINE | ID: mdl-35203528

ABSTRACT

Metformin (1,1-dimethylbiguanide hydrochloride) is the most commonly used drug to treat type II diabetic patients. It is believed that this drug has several other beneficial effects, such as anti-inflammatory and anticancer effects. Here, we wanted to evaluate the effect of metformin on the production of reactive oxygen species (ROS) by human macrophages. Macrophages are generated in vivo from circulating monocytes depending on the local tissue environment. In vitro proinflammatory macrophages (M1) and anti-inflammatory macrophages (M2) can be generated by culturing monocytes in the presence of different cytokines, such as GM-CSF or M-CSF, respectively. We show that metformin selectively inhibited human monocyte differentiation into proinflammatory macrophages (M1) without inhibiting their differentiation into anti-inflammatory macrophages (M2). Moreover, we demonstrate that, in response to LPS, M2 macrophages produced ROS, which could be very harmful for nearby tissues, and metformin inhibited this process. Interestingly, metformin with LPS induced activation of the adenosine-monophosphate-activated protein kinase (AMPK) and pharmacological activation of AMPK by AICAR, a known AMPK activator, decreased ROS production, whereas the deletion of AMPK in mice dramatically enhanced ROS production in different types of immune cells. These results suggest that metformin exhibits anti-inflammatory effects by inhibiting the differentiation of human monocytes into M1 macrophages and by limiting ROS production by macrophages via the activation of AMPK.

3.
Cell Mol Gastroenterol Hepatol ; 13(4): 1073-1093, 2022.
Article in English | MEDLINE | ID: mdl-35031518

ABSTRACT

BACKGROUND & AIMS: NADPH oxidase 1 (NOX1) has emerged as a prime regulator of intestinal mucosa immunity and homeostasis. Dysregulation of NOX1 may cause inflammatory bowel disease (IBD). It is not clear how NOX1 is regulated in vivo under inflammatory conditions. We studied the role of CK2 in this process. METHODS: The NOX1 organizer subunit, NADPH oxidase organizer 1 (NOXO1), was immunoprecipitated from cytokine-treated colon epithelial cells, and bound proteins were identified by mass spectrometry analysis. Sites on NOXO1 phosphorylated by CK2 were identified by nanoscale liquid chromatography coupled to tandem mass spectrometry. NOX1 activity was determined in colon epithelial cells and colonoids in the presence or absence of CX-4945, a CK2 specific inhibitor. Acute colitis was induced by administration of trinitrobenzenesulfonic acid in mice treated or not with CX-4945. Colon tissues were analyzed by histologic examination, quantitative polymerase chain reaction, and Western blots. CK2 activity, markers of inflammation, and oxidative stress were assessed. RESULTS: We identified CK2 as a major partner of NOXO1 in colon epithelial cells under inflammatory conditions. CK2 directly binds NOXO1 at the C-terminus containing the Phox homology domain and phosphorylates NOXO1 on several sites. CX-4945 increased ROS generation by NOX1 in human colon epithelial cells and organoids. Strikingly, CK2 activity was reduced in trinitrobenzenesulfonic acid-induced acute colitis, and CX-4945 exacerbated colitis inflammation as shown by increased levels of CXCL1, ROS generation, lipid peroxidation, and colon damage. CONCLUSIONS: The ubiquitous protein kinase CK2 limits NOX1 activity via NOXO1 binding and phosphorylation in colonic epithelial cells and lessens experimental colitis. Loss of CK2 activity during acute colitis results in excessive ROS production, contributing to the pathogenesis. Strategies to activate CK2 could be an effective novel therapeutic approach in IBD.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Animals , Casein Kinase II/adverse effects , Colitis/chemically induced , Inflammation , Mice , NADPH Oxidase 1/metabolism , Reactive Oxygen Species/metabolism , Trinitrobenzenesulfonic Acid/adverse effects
5.
J Exp Med ; 216(11): 2669-2687, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31492810

ABSTRACT

Neutrophils produce high levels of reactive oxygen species (ROS) by NADPH oxidase that are crucial for host defense but can lead to tissue injury when produced in excess. We previously described that proliferating cell nuclear antigen (PCNA), a nuclear scaffolding protein pivotal in DNA synthesis, controls neutrophil survival through its cytosolic association with procaspases. We herein showed that PCNA associated with p47phox, a key subunit of NADPH oxidase, and that this association regulated ROS production. Surface plasmon resonance and crystallography techniques demonstrated that the interdomain-connecting loop of PCNA interacted directly with the phox homology (PX) domain of the p47phox. PCNA inhibition by competing peptides or by T2AA, a small-molecule PCNA inhibitor, decreased NADPH oxidase activation in vitro. Furthermore, T2AA provided a therapeutic benefit in mice during trinitro-benzene-sulfonic acid (TNBS)-induced colitis by decreasing oxidative stress, accelerating mucosal repair, and promoting the resolution of inflammation. Our data suggest that targeting PCNA in inflammatory neutrophils holds promise as a multifaceted antiinflammatory strategy.


Subject(s)
Cytosol/metabolism , NADPH Oxidase 2/metabolism , NADPH Oxidases/metabolism , Neutrophils/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Colitis/chemically induced , Colitis/prevention & control , Enzyme Activation/drug effects , Female , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidases/genetics , Protein Binding , Reactive Oxygen Species/metabolism , Small Molecule Libraries/pharmacology , Trinitrobenzenesulfonic Acid
6.
Mucosal Immunol ; 12(1): 117-131, 2019 01.
Article in English | MEDLINE | ID: mdl-30279516

ABSTRACT

Inflammatory bowel disease (IBD) is characterized by severe and recurrent inflammation of the gastrointestinal tract, associated with altered patterns of cytokine synthesis, excessive reactive oxygen species (ROS) production, and high levels of the innate immune protein, lipocalin-2 (LCN-2), in the mucosa. The major source of ROS in intestinal epithelial cells is the NADPH oxidase NOX1, which consists of the transmembrane proteins, NOX1 and p22PHOX, and the cytosolic proteins, NOXO1, NOXA1, and Rac1. Here, we investigated whether NOX1 activation and ROS production induced by key inflammatory cytokines in IBD causally affects LCN-2 production in colonic epithelial cells. We found that the combination of TNFα and IL-17 induced a dramatic upregulation of NOXO1 expression that was dependent on the activation of p38MAPK and JNK1/2, and resulted into an increase of NOX1 activity and ROS production. NOX1-derived ROS drive the expression of LCN-2 by controlling the expression of IκBζ, a master inducer of LCN-2. Furthermore, LCN-2 production and colon damage were decreased in NOX1-deficient mice during TNBS-induced colitis. Finally, analyses of biopsies from patients with Crohn's disease showed increased JNK1/2 activation, and NOXO1 and LCN-2 expression. Therefore, NOX1 might play a key role in mucosal immunity and inflammation by controlling LCN-2 expression.


Subject(s)
Colitis/immunology , Colon/metabolism , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/metabolism , Lipocalin-2/metabolism , NADPH Oxidase 1/metabolism , Reactive Oxygen Species/metabolism , Animals , Cells, Cultured , Colitis/chemically induced , Colon/pathology , Cytochrome b Group/genetics , Cytokines/metabolism , Gene Expression Regulation , Humans , Immunity, Innate , Interleukin-17/metabolism , Intestinal Mucosa/pathology , Lipocalin-2/genetics , Mice , Mice, Knockout , NADPH Oxidase 1/genetics , NADPH Oxidases/genetics , RNA, Small Interfering/genetics , Signal Transduction , Trinitrobenzenesulfonic Acid/toxicity , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Blood ; 130(15): 1734-1745, 2017 10 12.
Article in English | MEDLINE | ID: mdl-28830888

ABSTRACT

Dendritic cells (DCs) are a heterogeneous population of professional antigen-presenting cells and are key cells of the immune system, acquiring different phenotypes in accordance with their localization during the immune response. A subset of inflammatory DCs is derived from circulating monocytes (Mo) and has a key role in inflammation and infection. The pathways controlling Mo-DC differentiation are not fully understood. Our objective was to investigate the possible role of nicotinamide adenine dinucleotide phosphate reduced form oxidases (NOXs) in Mo-DC differentiation. In this study, we revealed that Mo-DC differentiation was inhibited by NOX inhibitors and reactive oxygen species scavengers. We show that the Mo-DC differentiation was dependent on p22phox, and not on gp91phox/NOX2, as shown by the reduced Mo-DC differentiation observed in chronic granulomatous disease patients lacking p22phox. Moreover, we revealed that NOX5 expression was strongly increased during Mo-DC differentiation, but not during Mo-macrophage differentiation. NOX5 was expressed in circulating myeloid DC, and at a lower level in plasmacytoid DC. Interestingly, NOX5 was localized at the outer membrane of the mitochondria and interacted with p22phox in Mo-DC. Selective inhibitors and small interfering RNAs for NOX5 indicated that NOX5 controlled Mo-DC differentiation by regulating the JAK/STAT/MAPK and NFκB pathways. These data demonstrate that the NOX5-p22phox complex drives Mo-DC differentiation, and thus could be critical for immunity and inflammation.


Subject(s)
Cell Differentiation , Dendritic Cells/cytology , Membrane Proteins/metabolism , Monocytes/cytology , NADPH Oxidases/metabolism , Cell Differentiation/drug effects , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Humans , Membrane Glycoproteins/metabolism , Membrane Proteins/antagonists & inhibitors , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Models, Biological , Monocytes/drug effects , Monocytes/metabolism , NADPH Oxidase 2 , NADPH Oxidase 5 , NADPH Oxidases/antagonists & inhibitors , NF-kappa B/metabolism , Protein Binding/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
8.
BMC Complement Altern Med ; 16(1): 433, 2016 Nov 03.
Article in English | MEDLINE | ID: mdl-27809835

ABSTRACT

BACKGROUND: Anvillea garcinii Coss. & Durieu (Anv) plant is used as a traditional North African medicine against several diseases associated with inflammation. At inflammatory sites, reactive oxygen species (ROS) produced in excess by activated phagocyte NADPH oxidase (NOX2) can accentuate inflammatory responses. Thus, we investigated if Anv-water soluble polysaccharides could modulate primary human neutrophil oxidative burst in vitro. METHODS: Human neutrophils were isolated from fresh whole blood and O2.- generation was measured by cytochrome c reduction assays. Western blots were used to analyse the translocation of PKC, p47phox (a key component of NOX2 activity) to neutrophil plasma membrane. Also, myeloperoxidase (MPO) release in the extracellular medium was studied by western blots. Flow cytometric analysis was used to detect CD11b membrane expression. RESULTS: Water soluble polysaccharides from Anv dose-dependently inhibited N-formyl-methionyl-leucyl-phenylalanine (fMLF)- and phorbol myristate acetate (PMA)-induced O2.- generation by human neutrophils. Moreover, Anv-polysaccharides strongly inhibited PMA-induced PKCß and p47phox translocation to membranes and p47phox phosphorylation on Ser328, a main PKC target. In contrast, polysaccharides extract from Zygophyllum gaetulum plant, which is also used as a traditional North African medicine against inflammatory diseases, was ineffective on this PKCß-p47phox pathway. Further, Anv inhibited important neutrophil degranulation markers corresponding to myeloperoxidase (MPO) release and CD11b membrane expression. CONCLUSION: The process of down-regulating NADPH oxidase by polysaccharides extracts from Anv provides new insights into the mechanism of Anv's anti-inflammatory actions.


Subject(s)
Asteraceae/chemistry , Neutrophils/drug effects , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Plants, Medicinal/chemistry , Africa, Northern , Cell Degranulation/drug effects , Cells, Cultured , Humans , Leukocytes, Mononuclear/drug effects , Plant Extracts/chemistry
9.
Immunol Rev ; 273(1): 180-93, 2016 09.
Article in English | MEDLINE | ID: mdl-27558335

ABSTRACT

Neutrophils are the major circulating white blood cells in humans. They play an essential role in host defense against pathogens. In healthy individuals, circulating neutrophils are in a dormant state with very low efficiency of capture and arrest on the quiescent endothelium. Upon infection and subsequent release of pro-inflammatory mediators, the vascular endothelium signals to circulating neutrophils to roll, adhere, and cross the endothelial barrier. Neutrophils migrate toward the infection site along a gradient of chemo-attractants, then recognize and engulf the pathogen. To kill this pathogen entrapped inside the vacuole, neutrophils produce and release high quantities of antibacterial peptides, proteases, and reactive oxygen species (ROS). The robust ROS production is also called 'the respiratory burst', and the NADPH oxidase or NOX2 is the enzyme responsible for the production of superoxide anion, leading to other ROS. In vitro, several soluble and particulate agonists induce neutrophil ROS production. This process can be enhanced by prior neutrophil treatment with 'priming' agents, which alone do not induce a respiratory burst. In this review, we will describe the priming process and discuss the beneficial role of controlled neutrophil priming in host defense and the detrimental effect of excessive neutrophil priming in inflammatory diseases.


Subject(s)
Immunity, Innate , Inflammation/immunology , Neutrophil Activation , Neutrophils/physiology , Respiratory Burst , Animals , Cell Communication , Humans , Reactive Oxygen Species/metabolism , Transendothelial and Transepithelial Migration
10.
Biochem Pharmacol ; 104: 62-73, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26774451

ABSTRACT

Polymorphonuclear neutrophils are key players in host defense against pathogens through the robust production of superoxide anion by the NADPH oxidase and the release of antibacterial proteins from granules. However, inappropriate release of these agents in the extracellular environment induces severe tissue injury, thereby contributing to the physiopathology of acute and chronic inflammatory disorders. Many studies have been carried out to identify molecules capable of inhibiting phagocyte functions, in particular superoxide anion production, for therapeutic purposes. In the present study, we show that thymoquinone (TQ), the major component of the volatile oil from Nigella sativa (black cumin) seeds strongly inhibits fMLF-induced superoxide production and granules exocytosis in neutrophils. The inhibition of superoxide anion was not due to a scavenger effect, as TQ did not inhibit superoxide anion produced by the xanthine/xanthine oxidase system. Interestingly, TQ impaired the phosphorylation on Ser-304 and Ser-328 of p47(PHOX), a cytosolic subunit of the NADPH oxidase. TQ also attenuated specific and azurophilic granule exocytosis in fMLF-stimulated neutrophils as evidenced by decreased cell surface expression of gp91(PHOX) and CD11b, and release of myeloperoxidase. Furthermore, both the PKC and MAPK pathways, which are involved in p47(PHOX) phosphorylation and granules exocytosis, respectively, were inhibited by TQ in fMLF-stimulated neutrophils. Finally, in a model of pleurisy induced by λ-carrageenan in rats, TQ reduced neutrophil accumulation in the pleural space, showing that it not only inhibits PMN functions in vitro, but also exhibits anti-inflammatory properties in vivo. Thus, TQ possesses promising anti-inflammatory therapeutic potential.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Benzoquinones/pharmacology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Nigella sativa/chemistry , Adult , Animals , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/therapeutic use , Benzoquinones/isolation & purification , Benzoquinones/therapeutic use , Blotting, Western , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/enzymology , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Humans , Male , NADP/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Pleurisy/drug therapy , Pleurisy/immunology , Pleurisy/metabolism , Rats, Sprague-Dawley , Superoxides/antagonists & inhibitors , Superoxides/metabolism
11.
Inflamm Bowel Dis ; 21(5): 1018-26, 2015 May.
Article in English | MEDLINE | ID: mdl-25822013

ABSTRACT

BACKGROUND: Increased reactive oxygen species (ROS) production is associated with inflamed ileal lesions in Crohn's disease colonized by pathogenic adherent-invasive Escherichia coli LF82. We investigated whether such ileal bacteria can modulate ROS production by epithelial cells, thus impacting on inflammation and mucin expression. METHODS: Ileal bacteria from patients with Crohn's disease were incubated with cultured epithelial T84 cells, and ROS production was assayed using the luminol-amplified chemiluminescence method. The gentamicin protection assay was used for bacterial invasion of T84 cell. The expression of NADPH oxidase (NOX) subunits, mucin, and IL-8 was analyzed by quantitative real-time PCR and Western blots. Involvement of NOX and ROS was analyzed using diphenyleneiodonium (DPI) and N-acetylcysteine (NAC). RESULTS: Among different bacteria tested, only LF82 induced an increase of ROS production by T84 cells in a dose-dependent manner. This response was inhibited by DPI and NAC. Heat- or ethanol-attenuated LF82 bacteria and the mutant LF82ΔFimA, which does not express pili type 1 and poorly adheres to epithelial cells, did not induce the oxidative response. The LF82-induced oxidative response coincides with its invasion in T84 cells, and both processes were inhibited by DPI. Also, we observed an increased expression of NOX1 and NOXO1 in response to LF82 bacteria versus the mutant LF82ΔFimA. Furthermore, LF82 inhibited mucin gene expression (MUC2 and MUC5AC) in T84 cells while increasing the chemotactic IL-8 expression, both in a DPI-sensitive manner. CONCLUSIONS: Adherent-invasive E. coli LF82 induced ROS production by intestinal NADPH oxidase and altered mucin and IL-8 expression, leading to perpetuation of inflammatory lesions in Crohn's disease.


Subject(s)
Epithelial Cells/metabolism , Escherichia coli/pathogenicity , Ileum/metabolism , Intestinal Mucosa/metabolism , Mucin 5AC/metabolism , Mucin-2/metabolism , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Humans , Ileum/immunology , Ileum/microbiology , Immunoenzyme Techniques , Interleukin-8/genetics , Interleukin-8/metabolism , Intestines/immunology , Intestines/microbiology , Mucin 5AC/genetics , Mucin-2/genetics , NADPH Oxidase 1 , NADPH Oxidases/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
12.
Inflamm Bowel Dis ; 21(1): 198-207, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25208104

ABSTRACT

Advanced mucosal healing (MH) after intestinal mucosal inflammation coincides with sustained clinical remission and reduced rates of hospitalization and surgical resection, explaining why MH is increasingly considered as a full therapeutic goal and as an endpoint for clinical trials. Intestinal MH is a complex phenomenon viewed as a succession of steps necessary to restore tissue structure and function. These steps include epithelial cell migration and proliferation, cell differentiation, restoration of epithelial barrier functions, and modulation of cell apoptosis. Few clinical studies have evaluated the needs for specific macronutrients and micronutrients and their effects on intestinal MH, most data having been obtained from animal and cell studies. These data suggest that supplementation with specific amino acids including arginine, glutamine, glutamate, threonine, methionine, serine, proline, and the amino acid-derived compounds, polyamines can favorably influence MH. Short-chain fatty acids, which are produced by the microbiota from undigested polysaccharides and protein-derived amino acids, also exert beneficial effects on the process of intestinal MH in experimental models. Regarding supplementation with lipids, although the effects of ω-3 and ω-6 fatty acids remain controversial, endogenous prostaglandin synthesis seems to be necessary for MH. Finally, among micronutrients, several vitamin and mineral deficiencies with different frequencies have been observed in patients with inflammatory bowel diseases and supplementation with some of them (vitamin A, vitamin D3, vitamin C, and zinc) are presumed to favor MH. Future work, including clinical studies, should evaluate the efficiency of supplementation with combination of dietary compounds as adjuvant nutritional intervention for MH of the inflamed intestinal mucosa.


Subject(s)
Dietary Supplements , Inflammatory Bowel Diseases/diet therapy , Intestinal Mucosa/drug effects , Wound Healing , Humans
13.
Inflamm Bowel Dis ; 19(13): 2895-905, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24193156

ABSTRACT

BACKGROUND: Mucosal healing (MH) decreases the relapse risk in patients with inflammatory bowel disease, but the role of dietary supplementation in this process has been poorly investigated. Here, we investigated the effect of an amino acid mixture supplement on rat MH. METHODS: Colitis was induced using 5% of dextran sodium sulfate for 6 days. Then, rats received a mixture of threonine (0.50 g/d), methionine (0.31 g/d), and monosodium glutamate (0.57 g/d) or an isonitrogenous amount of alanine (control group). Colons were recovered after colitis induction and after dietary supplementation for measuring colon characteristics, myeloperoxidase, cytokine gene expression, glutathione content, protein synthesis rate, and for histological analysis. Short-chain fatty acids were measured in the colonic content. RESULTS: Colitis induction resulted in anorexia, thickening and shortening of the colon, and ulceration. Colonic cytokine expression and neutrophil infiltration were increased. An increased amount of water and a decreased amount of butyrate, propionate, and acetate were measured in the colonic content. Supplementation with the amino acid mixture coincided with a reduced protein synthesis rate in the colon compatible with the observed increased colonic MH. Mucosal regeneration/re-epithelialization was visible within 3 days after colitis induction at a time when mucosal inflammation was severe. Histological analysis revealed an increased regeneration/re-epithelialization after 10-day supplementation. In contrast, the spontaneous resolution of inflammation was not affected by the supplementation. CONCLUSIONS: Amino acid supplementation ameliorates colonic MH but not mucosal inflammatory status. Our data sustain the use of adjuvant dietary intervention on initiated intestinal MH.


Subject(s)
Amino Acids/administration & dosage , Colitis/drug therapy , Colon/drug effects , Intestinal Mucosa/drug effects , Animals , Colitis/chemically induced , Colitis/metabolism , Dextran Sulfate/toxicity , Disease Models, Animal , Glutathione/metabolism , Immunoenzyme Techniques , Male , Peroxidase/metabolism , Rats , Rats, Wistar
14.
Am J Blood Res ; 2(3): 187-93, 2012.
Article in English | MEDLINE | ID: mdl-23119229

ABSTRACT

The phagocyte NADPH oxidase (NOX2) is known to be expressed in Epstein-Barr virus (EBV)-transformed human B lymphocytes. Phosphorylation of the NOX2 cytosolic subunit p47phox is required for phorbol myristate acetate (PMA)-induced NOX2 activation in EBV-transformed B lymphocytes, however the role of this process in receptor-mediated NOX2 activation is not known. Here, we used pansorbin which acts by cross linking cell surface IgG and transfected cells with mutated p47phox to address if the phosphorylation of this subunit is required for receptor-mediated NOX2 activation. We show that pansorbin induced NOX2 activation in a time and concentration-dependent manner, albeit at levels only of 20% of those induced by PMA. GF109203X, a PKC selective inhibitor, inhibited pansorbin as well as PMA-induced NOX2 activation. Using specific anti-phospho serine antibodies we showed that pansorbin induced p47phox phosphorylation on Ser304, 315, 320, 328, and 345 and kinetics of these phosphorylations preceed NOX2 activation. To determine whether the phosphorylation of p47phox is required for pansorbin-induced NOX2 activation, we transfected EBV-transformed lymphocytes deficent in p47phox with a plasmid expressing wild type p47phox or p47phox with all the phosphorylated serines mutated to alanines, p47phoxS(303-379)A. Results show that pansorbin-induced NOX2 activation was greatly decreased in lymphocytes expressing the mutant as compared to the wild-type p47phox. These results show that pansorbin induced p47phox phosphorylation on multiple sites in EBV-transformed B lymphocytes and this process is required for pansorbin-induced NADPH oxidase activation in these cells.

15.
Am J Pathol ; 180(2): 682-92, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22119038

ABSTRACT

Adiponectin (Acrp30) belongs to the family of C1q/tumor necrosis factor α (TNFα)-related proteins. Acrp30 circulates as multimers of high, middle, and low molecular weight. In this study, we detected Acrp30 and its globular fragment (gAcrp30) in synovial fluid from rheumatoid arthritis patients. Intriguingly, the LMW form was more abundant in synovial fluid than in serum from both rheumatoid arthritis patients and healthy subjects. We also investigated the effects of Acrp30 and gAcrp30 on reactive oxygen species (ROS) production via the phagocytic NADPH oxidase. Acrp30 inhibited fMLF-induced ROS production by human phagocytes, whereas gAcrp30 enhanced it. gAcrp30's effect is additive with TNFα, whereas Acrp30 inhibited TNFα-induced priming. gAcrp30 enhanced NOX-2 expression at the plasma membrane, with a concomitant increase in p47(phox) phosphorylation. Selective inhibitors of p38 mitogen-activated protein kinase (MAPK) and extracellular signal-regulated kinase 1 (ERK1)/2 abrogated p47(phox) phosphorylation by gAcrp30. In contrast, p47(phox) phosphorylation was inhibited by Acrp30 in association with increased AMP-activated protein kinase (AMPK) phosphorylation in phagocytes. These results suggest that human phagocyte ROS production is regulated by different mechanisms selective for Acrp30 versus gAcrp30. An imbalance between gAcrp30 and higher molecular weight isoforms of Acrp30 might contribute to chronic inflammation by regulating NADPH oxidase.


Subject(s)
Adiponectin/physiology , Arthritis, Rheumatoid/metabolism , Phagocytes/metabolism , Reactive Oxygen Species/metabolism , Respiratory Burst/physiology , Adiponectin/metabolism , CD11b Antigen/metabolism , Humans , MAP Kinase Signaling System/physiology , Monocytes/metabolism , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , NADPH Oxidases/metabolism , Neutrophils/metabolism , Phosphorylation , Protein Isoforms , Protein Transport/physiology , Synovial Fluid/chemistry , p38 Mitogen-Activated Protein Kinases/metabolism
16.
PLoS One ; 4(7): e6458, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19649246

ABSTRACT

BACKGROUND: Neutrophils play a major role in inflammation by releasing large amounts of ROS produced by NADPH-oxidase and myeloperoxidase (MPO). The proinflammatory cytokine TNFalpha primes ROS production through phosphorylation of the NADPH-oxidase subunit p47phox on Ser345. Conventional anti-inflammatory therapies remain partially successful and may have side effects. Therefore, regulation of neutrophil activation by natural dietary components represents an alternative therapeutic strategy in inflammatory diseases such as inflammatory bowel diseases. The aim of this study was to assess the effect of punicic acid, a conjugated linolenic fatty acid from pomegranate seed oil on TNFalpha-induced neutrophil hyperactivation in vitro and on colon inflammation in vivo. METHODOLOGY AND PRINCIPAL FINDINGS: We analyzed the effect of punicic acid on TNFalpha-induced neutrophil upregulation of ROS production in vitro and on TNBS-induced rat colon inflammation. Results show that punicic acid inhibited TNFalpha-induced priming of ROS production in vitro while preserving formyl-methionyl-leucyl-phenylalanine (fMLP)-induced response. This effect was mediated by the inhibition of Ser345-p47phox phosphorylation and upstream kinase p38MAPK. Punicic acid also inhibited fMLP- and TNFalpha+fMLP-induced MPO extracellular release from neutrophils. In vivo experiments showed that punicic acid and pomegranate seed oil intake decreased neutrophil-activation and ROS/MPO-mediated tissue damage as measured by F2-isoprostane release and protected rats from TNBS-induced colon inflammation. CONCLUSIONS/SIGNIFICANCE: These data show that punicic acid exerts a potent anti-inflammatory effect through inhibition of TNFalpha-induced priming of NADPH oxidase by targeting the p38MAPKinase/Ser345-p47phox-axis and MPO release. This natural dietary compound may provide a novel alternative therapeutic strategy in inflammatory diseases such as inflammatory bowel diseases.


Subject(s)
Colitis/prevention & control , Linolenic Acids/pharmacology , Lymphocyte Activation/drug effects , Neutrophils/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , NADPH Oxidases/metabolism , Neutrophils/metabolism , Phosphorylation , Rats , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Diabetes ; 58(9): 2032-8, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19502416

ABSTRACT

OBJECTIVE: An increased expression of RELM-beta (resistin-like molecule-beta), a gut-derived hormone, is observed in animal models of insulin resistance/obesity and intestinal inflammation. Intestinal sugar absorption is modulated by dietary environment and hormones/cytokines. The aim of this study was to investigate the effect of RELM-beta on intestinal glucose absorption. RESEARCH DESIGN AND METHODS: Oral glucose tolerance test was performed in mice and rats in the presence and the absence of RELM-beta. The RELM-beta action on glucose transport in rat jejunal sacs, everted rings, and mucosal strips was explored as well as downstream kinases modulating SGLT-1 and GLUT2 glucose transporters. RESULTS: Oral glucose tolerance test carried out in rodents showed that oral administration of RELM-beta increased glycemia. Studies in rat jejunal tissue indicated that mucosal RELM-beta promoted absorption of glucose from the gut lumen. RELM-beta had no effect on paracellular mannitol transport, suggesting a transporter-mediated transcellular mechanism. In studies with jejunal mucosa mounted in Ussing chamber, luminal RELM-beta inhibited SGLT-1 activity in line with a diminished SGLT-1 abundance in brush border membranes (BBMs). Further, the potentiating effect of RELM-beta on jejunal glucose uptake was associated with an increased abundance of GLUT2 at BBMs. The effects of RELM-beta were associated with an increased amount of protein kinase C betaII in BBMs and an increased phosphorylation of AMP-activated protein kinase (AMPK). CONCLUSIONS: The regulation of SGLT-1 and GLUT2 by RELM-beta expands the role of gut hormones in short-term AMPK/protein kinase C mediated control of energy balance.


Subject(s)
Glucose Transporter Type 2/metabolism , Glucose/pharmacokinetics , Hormones, Ectopic/metabolism , Jejunum/metabolism , Sodium-Glucose Transporter 1/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , Glucose Tolerance Test , Hormones, Ectopic/pharmacology , Intercellular Signaling Peptides and Proteins , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Jejunum/drug effects , Male , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Kinase C/metabolism , Protein Kinase C beta , Rats , Rats, Wistar , Recombinant Proteins/pharmacology , Sodium/metabolism
18.
Exp Mol Med ; 41(4): 217-25, 2009 Apr 30.
Article in English | MEDLINE | ID: mdl-19372727

ABSTRACT

Phagocytes such as neutrophils play a vital role in host defense against microbial pathogens. The anti-microbial function of neutrophils is based on the production of superoxide anion (O2 -), which generates other microbicidal reactive oxygen species (ROS) and release of antimicrobial peptides and proteins. The enzyme responsible for O2 - production is called the NADPH oxidase or respiratory burst oxidase. This multicomponent enzyme system is composed of two trans- membrane proteins (p22phox and gp91phox, also called NOX2, which together form the cytochrome b558) and four cytosolic proteins (p47phox, p67phox, p40phox and a GTPase Rac1 or Rac2), which assemble at membrane sites upon cell activation. NADPH oxidase activation in phagocytes can be induced by a large number of soluble and particulate agents. This process is dependent on the phosphorylation of the cytosolic protein p47phox. p47phox is a 390 amino acids protein with several functional domains: one phox homology (PX) domain, two src homology 3 (SH3) domains, an auto-inhibitory region (AIR), a proline rich domain (PRR) and has several phosphorylated sites located between Ser303 and Ser379. In this review, we will describe the structure of p47phox, its phosphorylation and discuss how these events regulate NADPH oxidase activation.


Subject(s)
Disease , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Phagocytes/metabolism , Enzyme Activation , Humans , Membrane Glycoproteins/chemistry , NADPH Oxidase 2 , NADPH Oxidases/chemistry , NADPH Oxidases/genetics , Phagocytes/cytology , Phosphorylation , Protein Conformation
19.
Inflamm Bowel Dis ; 14(7): 931-41, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18300276

ABSTRACT

BACKGROUND: Resistin and resistin-like molecule (RELM)beta comprise a novel class of cysteine-rich proteins secreted into the circulation implicated in hepatic insulin resistance and inflammation. RELMbeta is specifically produced by intestinal goblet cells but regulation of its expression and much of its local function are not elucidated. RELMbeta has been suggested to regulate colonic inflammation susceptibility, which is dependent on the mucosal barrier integrity. METHODS: In this work we explored the physiopathological role of RELMbeta in the colon. Among agents tested, carbachol and gastrin were strong inhibitors of RELMbeta mRNA accumulation. We examined the effect of recombinant RELMbeta on mucin secretion by human mucus-secreting HT29-Cl.16E cells in culture and by mouse colonic goblet cells in vivo. RESULTS: RELMbeta upregulated MUC2 and M1/MUC5AC gene expression in HT29-Cl.16E cells. RELMbeta enhanced M1/MUC5AC secretion by human colonic HT29-Cl.16E cells and MUC2 secretion by murine intestinal goblet cells. RELMbeta exerted its action exclusively on the apical side of HT29-Cl.16E cells, in agreement with its luminal mucosecretagogue effect in mice. Its action required calcium, protein kinase C, tyrosine kinases, and extracellular-regulated protein kinase activities and was synergized by carbachol. An intracolonic RELMbeta challenge was performed in the trinitrobenzene sulfonic acid (TNBS)-murine model of colitis and macroscopic and histological scores were monitored. The macroscopic and histopathological severity of TNBS-induced colitis was significantly attenuated by RELMbeta pretreatment. CONCLUSIONS: A direct participation in maintaining the mucosal defense barrier can be ascribed to RELMbeta in line with a regulatory role in intestinal inflammation.


Subject(s)
Colitis/physiopathology , Hormones, Ectopic/physiology , Intestinal Mucosa/metabolism , Mucus/metabolism , Trinitrobenzenesulfonic Acid , Animals , Blotting, Western , Calcium/physiology , Carbachol/pharmacology , Cells, Cultured , Colitis/chemically induced , Colitis/pathology , Gastrins/pharmacology , Gene Expression , Goblet Cells/metabolism , Hormones, Ectopic/genetics , Hormones, Ectopic/pharmacology , Humans , Intercellular Signaling Peptides and Proteins , Intestines , Mice , Mucin 5AC , Mucin-2 , Mucins/genetics , Protein Kinase C/physiology , Protein Kinases/physiology , Protein-Tyrosine Kinases/physiology , RNA, Messenger/drug effects , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
20.
FASEB J ; 20(9): 1504-6, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16720733

ABSTRACT

Interleukin-10 (IL-10) exerts its anti-inflammatory properties by down-regulating polymorphonuclear neutrophil (PMN) functions such as reactive oxygen species (ROS) production via NADPH oxidase. The molecular mechanisms underlying this process are unclear. Partial phosphorylation of the NADPH oxidase cytosolic component p47(PHOX) induced by proinflammatory cytokines, such as granulocyte-macrophage colony stimulating factor (GM-CSF) and tumor necrosis factor (TNF)-alpha, is essential for priming ROS production by PMN. The aim of this study was to determine whether IL-10 inhibits GM-CSF- and TNFalpha-induced p47(PHOX) phosphorylation and to investigate the molecular mechanisms involved in this effect. We found that IL-10 selectively inhibited GM-CSF- but not TNFalpha-induced p47PHOX phosphorylation in a concentration-dependent manner. As GM-CSF-induced p47PHOX phosphorylation is mediated by extracellular signal-regulated kinase 1/2 (ERK1/2), we tested the effect of IL-10 on this pathway. We found that IL-10 inhibited GM-CSF-induced ERK1/2 activity in an immunocomplex kinase assay. This inhibitory effect was confirmed by analyzing the phosphorylation status of the endogenous substrate of ERK1/2, p90RSK, in intact PMN. Furthermore, IL-10 decreased ROS production by adherent GM-CSF-treated PMN in keeping with the higher ROS production observed in whole blood from IL-10 knockout mice compared to their wild-type counterparts. Together, these results suggest that IL-10 inhibits GM-CSF-induced priming of ROS production by inhibiting p47PHOX phosphorylation through a decrease in ERK1/2 activity. This IL-10 effect could contribute to the tight regulation of NADPH oxidase activity at the inflammatory site.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interleukin-10/pharmacology , NADPH Oxidases/metabolism , Neutrophils/physiology , Respiratory Burst/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NADPH Oxidases/drug effects , Neutrophils/drug effects , Phosphorylation , Respiratory Burst/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...