Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 13 de 13
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Inflammation ; 47(1): 30-44, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-37603227

RÉSUMÉ

Activated microglia-induced inflammation in the hippocampus plays an important role in perioperative neurocognitive disorders. Previous studies have shown that sialic acid-binding immunoglobulin-like lectin 3 (hSiglec-3, ortholog of mouse Siglec-E) engagement in microglia and its glycan ligands on neurons contributes to inflammatory homeostasis through an endogenous negative regulation pathway. This study aimed to explore whether the glycan ligand alteration on neurons plays a role in sevoflurane-induced perioperative neurocognitive disorders. This study's data has shown that a slight Siglec-E ligands' expression decrease does not induce inflammation homeostasis disruption. We also demonstrated that the ligand level on neurons was decreased with age, and the reduced Siglec-E ligand expression on neurons caused via sevoflurane was induced by neuraminidase 1. Furthermore, this study has shown that the Siglec-E ligand expression decline caused by age and sevoflurane treatment could decrease the ligands' level, thus leading to inflammatory homeostasis disruption. This research provided a novel mechanism for perioperative neurocognitive disorder susceptibility in the elderly.


Sujet(s)
Inflammation , Lectines liant l'acide sialique apparentées aux immunoglobulines , Humains , Souris , Animaux , Sujet âgé , Lectines liant l'acide sialique apparentées aux immunoglobulines/métabolisme , Ligands , Sévoflurane/effets indésirables , Régulation négative , Inflammation/induit chimiquement , Inflammation/métabolisme , Neurones/métabolisme , Polyosides/métabolisme , Hippocampe/métabolisme
3.
Front Cell Neurosci ; 17: 1207540, 2023.
Article de Anglais | MEDLINE | ID: mdl-37492129

RÉSUMÉ

Multiple sclerosis is an inflammatory demyelinating disease of the central nervous system. Spontaneous restoration of myelin after demyelination occurs, but its efficiency declines during disease progression. Efficient myelin repair requires fine-tuning inflammatory responses by brain-resident microglia and infiltrating macrophages. Accordingly, promising therapeutic strategies aim at controlling inflammation to promote remyelination. Polysialic acid (polySia) is a polymeric glycan with variable chain lengths, presented as a posttranslational modification on select protein carriers. PolySia emerges as a negative regulator of inflammatory microglia and macrophage activation and has been detected on oligodendrocyte precursors and reactive astrocytes in multiple sclerosis lesions. As shown recently, polySia-modified proteins can also be released by activated microglia, and the intrinsically released protein-bound and exogenously applied free polySia were equally able to attenuate proinflammatory microglia activation via the inhibitory immune receptor Siglec-E. In this study, we explore polySia as a candidate substance for promoting myelin regeneration by immunomodulation. Lysophosphatidylcholine-induced demyelination of organotypic cerebellar slice cultures was used as an experimental model to analyze the impact of polySia with different degrees of polymerization (DP) on remyelination and inflammation. In lysophosphatidylcholine-treated cerebellar slice cultures, polySia-positive cells were abundant during demyelination but largely reduced during remyelination. Based on the determination of DP24 as the minimal polySia chain length required for the inhibition of inflammatory BV2 microglia activation, pools with short and long polySia chains (DP8-14 and DP24-30) were generated and applied to slice cultures during remyelination. Unlike DP8-14, treatment with DP24-30 significantly improved remyelination, increased arginase-1-positive microglia ratios, and reduced the production of nitric oxide in wildtype, but not in Siglec-E-deficient slice cultures. In vitro differentiation of oligodendrocytes was not affected by DP24-30. Collectively, these results suggest a beneficial effect of exogenously applied polySia DP24-30 on remyelination by Siglec-E-dependent microglia regulation.

4.
Cell Metab ; 34(8): 1088-1103.e6, 2022 08 02.
Article de Anglais | MEDLINE | ID: mdl-35921817

RÉSUMÉ

The molecular interactions that regulate chronic inflammation underlying metabolic disease remain largely unknown. Since the CD24-Siglec interaction regulates inflammatory response to danger-associated molecular patterns (DAMPs), we have generated multiple mouse strains with single or combined mutations of Cd24 or Siglec genes to explore the role of the CD24-Siglec interaction in metaflammation and metabolic disorder. Here, we report that the CD24-Siglec-E axis, but not other Siglecs, is a key suppressor of obesity-related metabolic dysfunction. Inactivation of the CD24-Siglec-E pathway exacerbates, while CD24Fc treatment alleviates, diet-induced metabolic disorders, including obesity, dyslipidemia, insulin resistance, and nonalcoholic steatohepatitis (NASH). Mechanistically, sialylation-dependent recognition of CD24 by Siglec-E induces SHP-1 recruitment and represses metaflammation to protect against metabolic syndrome. A first-in-human study of CD24Fc (NCT02650895) supports the significance of this pathway in human lipid metabolism and inflammation. These findings identify the CD24-Siglec-E axis as an innate immune checkpoint against metaflammation and metabolic disorder and suggest a promising therapeutic target for metabolic disease.


Sujet(s)
Maladies métaboliques , Lectines liant l'acide sialique apparentées aux immunoglobulines , Animaux , Antigènes CD24/génétique , Antigènes CD24/métabolisme , Études cliniques comme sujet , Humains , Inflammation , Souris , Obésité , Phagocytose , Lectines liant l'acide sialique apparentées aux immunoglobulines/métabolisme
5.
Front Immunol ; 12: 626110, 2021.
Article de Anglais | MEDLINE | ID: mdl-33763070

RÉSUMÉ

TLR4 activates two distinct signaling pathways involving adaptors MyD88 and TRIF to produce proinflammatory cytokines and type-I interferon respectively. How Leishmania donovani suppresses these pathways is not well studied. We earlier reported, TLR4 is hypersialylated due to reduced membrane-bound neuraminidase (Neu1) on infected-macrophages. We hypothesized that such enhanced sialoglycoconjugates on host cells may modulate the interactions with siglecs- which are the inhibitory receptors. Here, we examined the impact of such sialylation on overall TLR4 activation both in murine cell line J774A.1 and primary bone marrow derived macrophages (BMDM). Supporting this hypothesis, we demonstrated siglec-E engages hypersialylated TLR4 during infection. Such sialic acids-siglec-E interaction enhanced siglec-E phosphorylation that mediated its strong association with SHP1/SHP2 and also upregulated their phosphorylation in both types of macrophages. Pre-treatment of parasites and host cells with neuraminidase reduced SHP1/SHP2 phosphorylation and triggered TLR4 activation respectively through enhanced nuclear translocation of p-65. Moreover, a reciprocal interplay between Neu1 and siglec-E differentially regulates MyD88- and TRIF-pathways through sialic acids on TLR4 as their common substrate during infection. Correspondingly, Neu1 overexpression enhanced MyD88-signaling while still suppressing TRIF-activation. However, silencing siglec-E specifically activated TRIF-signaling. Pro-inflammatory cytokines corresponding to MyD88 and TRIF pathways were also upregulated respectively. Additionally, Neu1 overexpression or siglec-E silencing prevented TLR4 ubiquitination and subsequent degradation by Triad3A. Neu1-overexpression and siglec-E-silencing together followed by infection activated both MyD88 and TRIF-signaling through their enhanced TLR4-association. This elevated the MyD88-specific cytokines and TRIF-mediated IRF3 and IFN-ß genes, thus upregulating the pro-inflammatory cytokines and nitric oxide levels and reduced anti-inflammatory cytokines. All these significantly inhibited parasite survival in macrophages thus demonstrating a previously unidentified dualistic regulation of TLR4signaling pathways activation through sialic acids by interplay of Neu1 and siglec-E during Leishmania infection.


Sujet(s)
Protéines adaptatrices du transport vésiculaire/métabolisme , Antigènes CD/métabolisme , Antigènes de différenciation des lymphocytes B/métabolisme , Leishmania donovani/pathogénicité , Leishmaniose viscérale/parasitologie , Macrophages/parasitologie , Facteur de différenciation myéloïde-88/métabolisme , Sialidase/métabolisme , Acides sialiques/métabolisme , Récepteur de type Toll-4/métabolisme , Animaux , Antigènes CD/génétique , Antigènes de différenciation des lymphocytes B/génétique , Lignée cellulaire , Cytokines/génétique , Cytokines/métabolisme , Modèles animaux de maladie humaine , Interactions hôte-pathogène , Immunité innée , Médiateurs de l'inflammation/métabolisme , Leishmania donovani/immunologie , Leishmaniose viscérale/immunologie , Leishmaniose viscérale/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Mesocricetus , Souris de lignée BALB C , Sialidase/génétique , Monoxyde d'azote/métabolisme , Phosphorylation , Transduction du signal
6.
J Biomed Sci ; 28(1): 5, 2021 Jan 05.
Article de Anglais | MEDLINE | ID: mdl-33397354

RÉSUMÉ

BACKGROUND: The accumulation of lipid-laden macrophages, foam cells, within sub-endothelial intima is a key feature of early atherosclerosis. Siglec-E, a mouse orthologue of human Siglec-9, is a sialic acid binding lectin predominantly expressed on the surface of myeloid cells to transduce inhibitory signal via recruitment of SH2-domain containing protein tyrosine phosphatase SHP-1/2 upon binding to its sialoglycan ligands. Whether Siglec-E expression on macrophages impacts foam cell formation and atherosclerosis remains to be established. METHODS: ApoE-deficient (apoE-/-) and apoE/Siglec-E-double deficient (apoE-/-/Siglec-E-/-) mice were placed on high fat diet for 3 months and their lipid profiles and severities of atherosclerosis were assessed. Modified low-density lipoprotein (LDL) uptake and foam cell formation in wild type (WT) and Siglec-E-/-- peritoneal macrophages were examined in vitro. Potential Siglec-E-interacting proteins were identified by proximity labeling in conjunction with proteomic analysis and confirmed by coimmunoprecipitation experiment. Impacts of Siglec-E expression and cell surface sialic acid status on oxidized LDL uptake and signaling involved were examined by biochemical assays. RESULTS: Here we show that genetic deletion of Siglec-E accelerated atherosclerosis without affecting lipid profile in apoE-/- mice. Siglec-E deficiency promotes foam cell formation by enhancing acetylated and oxidized LDL uptake without affecting cholesterol efflux in macrophages in vitro. By performing proximity labeling and proteomic analysis, we identified scavenger receptor CD36 as a cell surface protein interacting with Siglec-E. Further experiments performed in HEK293T cells transiently overexpressing Siglec-E and CD36 and peritoneal macrophages demonstrated that depletion of cell surface sialic acids by treatment with sialyltransferase inhibitor or sialidase did not affect interaction between Siglec-E and CD36 but retarded Siglec-E-mediated inhibition on oxidized LDL uptake. Subsequent experiments revealed that oxidized LDL induced transient Siglec-E tyrosine phosphorylation and recruitment of SHP-1 phosphatase in macrophages. VAV, a downstream effector implicated in CD36-mediated oxidized LDL uptake, was shown to interact with SHP-1 following oxidized LDL treatment. Moreover, oxidized LDL-induced VAV phosphorylation was substantially lower in WT macrophages comparing to Siglec-E-/- counterparts. CONCLUSIONS: These data support the protective role of Siglec-E in atherosclerosis. Mechanistically, Siglec-E interacts with CD36 to suppress downstream VAV signaling involved in modified LDL uptake.


Sujet(s)
Apolipoprotéines E/déficit , Athérosclérose/génétique , Antigènes CD36/métabolisme , Cellules spumeuses/métabolisme , Lectines liant l'acide sialique apparentées aux immunoglobulines/pharmacologie , Animaux , Athérosclérose/métabolisme , Souris
7.
Bone ; 143: 115665, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33007530

RÉSUMÉ

Regulation of osteoclast differentiation and function is a central element in bone homeostasis. While the role of soluble factors, such as cytokines, hormones and growth factors, in controlling osteoclast differentiation has been intensively characterized, the function of surface receptors is less well understood. Sialic acid-binding immunoglobulin-like lectin (Siglec)-9 and its murine homolog Siglec-E are sialic acid-recognizing inhibitory receptors from the CD33-related Siglec-family and mainly expressed on myeloid cells. We found Siglec-9 and Siglec-E to be expressed at all stages of human and murine osteoclastogenesis, respectively. Siglec-E knockout mice displayed lower bone mass despite unchanged osteoclast numbers and an increased bone formation rate. Ex vivo osteoclast assays using Siglec-E knockout cells or a blocking antibody against human Siglec-9 confirmed the suppressive effect of Siglec-9/Siglec-E on osteoclast function. Although osteoclast numbers were unchanged or even slightly decreased, the blockade/absence of Siglec-9/Siglec-E resulted in an augmented resorption activity of mature osteoclasts. This increased resorption activity was associated with enlarged actin rings. Together, our results suggest Siglec-9/Siglec-E to inhibit osteoclast activation independently from osteoclast differentiation and thereby propose a new mechanism for the control of local bone resorption.


Sujet(s)
Résorption osseuse , Ostéoclastes , Animaux , Différenciation cellulaire , Humains , Immunoglobulines , Protéines membranaires , Souris , Acide N-acétyl-neuraminique , Lectines liant l'acide sialique apparentées aux immunoglobulines
8.
Front Immunol ; 11: 332, 2020.
Article de Anglais | MEDLINE | ID: mdl-32184783

RÉSUMÉ

Pseudomonas aeruginosa (PA) is commonly associated with nosocomial and chronic infections of lungs. We have earlier demonstrated that an acidic sugar, sialic acid, is present in PA which is recognized and bound by sialic acid binding immunoglobulin type lectins (siglecs) expressed on neutrophils. Here, we have tried to gain a detailed insight into the immunosuppressive role of sialic acid-siglec interactions in macrophage-mediated clearance of sialylated PA (PA+Sia). We have demonstrated that PA+Sia shows enhanced binding (~1.5-fold) to macrophages due to additional interactions between sialic acids and siglec-E and exhibited more phagocytosis. However, internalization of PA+Sia is associated with a reduction in respiratory burst and increase in anti-inflammatory cytokines secretion which is reversed upon desialylation of the bacteria. Phagocytosis of PA+Sia is also associated with reduced intracellular calcium ion concentrations and altered calcium-dependent signaling which negatively affects phagosome maturation. Consequently, although more PA+Sia was localized in early phagosomes (Rab5 compartment), only fewer bacteria reach into the late phagosomal compartment (Rab7). Possibly, this leads to reduced phagosome lysosome fusion where reduced numbers of PA+Sia are trafficked into lysosomes, compared to PA-Sia. Thus, internalized PA+Sia remain viable and replicates intracellularly in macrophages. We have also demonstrated that such siglec-E-sialic acid interaction recruited SHP-1/SHP-2 phosphatases which modulate MAPK and NF-κB signaling pathways. Disrupting sialic acid-siglec-E interaction by silencing siglec-E in macrophages results in improved bactericidal response against PA+Sia characterized by robust respiratory burst, enhanced intracellular calcium levels and nuclear translocation of p65 component of NF-κB complex leading to increased pro-inflammatory cytokine secretion. Taken together, we have identified that sialic acid-siglec-E interactions is another pathway utilized by PA in order to suppress macrophage antimicrobial responses and inhibit phagosome maturation, thereby persisting as an intracellular pathogen in macrophages.


Sujet(s)
Poumon/immunologie , Macrophages/immunologie , Granulocytes neutrophiles/immunologie , Phagosomes/métabolisme , Infections à Pseudomonas/immunologie , Pseudomonas aeruginosa/physiologie , Infections de l'appareil respiratoire/immunologie , Animaux , Antigènes CD/métabolisme , Antigènes de différenciation des lymphocytes B/métabolisme , Calcium/métabolisme , Signalisation calcique , Humains , Échappement immunitaire , Tolérance immunitaire , Poumon/microbiologie , Souris , Acide N-acétyl-neuraminique/génétique , Acide N-acétyl-neuraminique/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Oxydoréduction , Phagocytose , Liaison aux protéines , Petit ARN interférent/génétique , Cellules THP-1
9.
Biomed Pharmacother ; 122: 109760, 2020 Feb.
Article de Anglais | MEDLINE | ID: mdl-31918287

RÉSUMÉ

AIMS: Siglec-E, the mouse ortholog of human Siglec-9, is an immunosuppressive cell surface receptor. Both Siglec-E and Siglec-9 are primarily found on neutrophils, macrophages, and monocytes. When Siglec-E binds to sialoglycan ligands in its extracellular environment, it halts the immune cells' inflammatory responses. In the present study, we aimed to investigate expression, mechanisms of action and regulation of Siglec-E ligands during vascular inflammation induced by E. coli lipopolysaccharides (LPS) in mouse aorta. METHODS: The distribution, molecular size and glycoprotein class of Siglec-E ligands on mouse aorta were determined, and the protein carrier of the ligands was identified. In vivo, the expression of Siglec-E ligands was detected after LPS treatment, with or without NF-κB inhibitor administration. In vitro, cultured primary mouse aortic endothelial cells (MAECs) were used to study changes in expression of Siglec-E ligands induced by LPS with or without NF-κB inhibitors. MAECs induced by LPS were co-cultured with macrophages and the effect of increased expression of Siglec-E ligands analyzed. RESULTS: Siglec-E ligands are O-linked sialoglycoproteins with molecular weights of 70-300 kDa and are distributed broadly on mouse aorta as well as on MAECs in vitro. In vivo, the expression of Siglec-E ligands was increased in mice aortas in response to LPS treatment in an NF-κB signaling pathway dependent manner. In MAECs, the expression of Siglec-E ligands was also increased by LPS via an NF-κB signaling pathway. Deleted in malignant brain tumors-1 was identified to be one of multiple protein carriers of Siglec-E ligands, and glycans of ligands involved in MAECs induced by LPS. Notably, co-incubation of macrophages with LPS-treated MAECs induced macrophage apoptosis and decreased macrophage phagocytosis, effects that were completely reversed by blocking Siglec-E binding to Siglec-E ligands. CONCLUSIONS: These data demonstrated that Siglec-E ligands were highly expressed in response to LPS-induced vascular inflammation and inhibited the immune response of macrophages, which may be a therapeutic strategy to interfere with vascular inflammation.


Sujet(s)
Antigènes CD/métabolisme , Antigènes de différenciation des lymphocytes B/métabolisme , Immunosuppresseurs/pharmacologie , Lipopolysaccharides/pharmacologie , Facteur de transcription NF-kappa B/métabolisme , Lectines liant l'acide sialique apparentées aux immunoglobulines/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Régulation positive/effets des médicaments et des substances chimiques , Animaux , Aorte/effets des médicaments et des substances chimiques , Aorte/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Escherichia coli/effets des médicaments et des substances chimiques , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Ligands , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Souris , Souris de lignée BALB C , Monocytes/effets des médicaments et des substances chimiques , Monocytes/métabolisme , Transduction du signal/physiologie , Activation de la transcription/effets des médicaments et des substances chimiques
10.
Int Arch Allergy Immunol ; 175(1-2): 5-15, 2018.
Article de Anglais | MEDLINE | ID: mdl-29306942

RÉSUMÉ

Severe asthma comprises only 5% of patients with asthma, but the burden it brings to the social health system accounts for more than half of all asthmatics. Clinical evidence shows that severe asthma is often linked to the recruitment and activation of neutrophils in the airways. However, the underlying molecular and immunological mechanisms of neutrophilia in severe asthma are not clear and currently available drugs exert only limited effects on neutrophilic inflammation. Great efforts are underway to address the mystery of neutrophilic inflammation in chronic respiratory disorders. Sialic acid-binding immunoglobulin-like lectins (Siglecs) are members of the immunoglobulin gene family. Of note, Siglec-9 is uniquely expressed by human neutrophils and monocytes, as well as a minor population of natural killer cells. Engaging this structure with antibodies or glycan ligands results in programmed cell death in human neutrophils. Intriguingly, the administration of Siglec-E antibody abolished the recruitment of neutrophils in mouse models of neutrophilic pulmonary inflammation in vivo. Given that neutrophils are probably a major culprit in the generation and perpetuation of inflammation, targeting Siglec-9 could be beneficial for the treatment of severe asthma, chronic obstructive pulmonary disease, and related pulmonary disorders characteristic of neutrophilia.


Sujet(s)
Anticorps monoclonaux/usage thérapeutique , Antigènes CD/métabolisme , Asthme/immunologie , Immunothérapie/méthodes , Granulocytes neutrophiles/métabolisme , Broncho-pneumopathie chronique obstructive/thérapie , Lectines liant l'acide sialique apparentées aux immunoglobulines/métabolisme , Animaux , Antigènes CD/immunologie , Antigènes de différenciation des lymphocytes B/immunologie , Asthme/thérapie , Mouvement cellulaire , Modèles animaux de maladie humaine , Humains , Souris , Granulocytes neutrophiles/immunologie , Broncho-pneumopathie chronique obstructive/immunologie
11.
J Biol Chem ; 292(3): 1029-1037, 2017 01 20.
Article de Anglais | MEDLINE | ID: mdl-27920204

RÉSUMÉ

CD33-related Siglecs are a family of proteins widely expressed on innate immune cells. Binding of sialylated glycans or other ligands triggers signals that inhibit or activate inflammation. Immunomodulation by Siglecs has been extensively studied, but relationships between structure and functions are poorly explored. Here we present new data relating to the structure and function of Siglec-E, the major CD33-related Siglec expressed on mouse neutrophils, monocytes, macrophages, and dendritic cells. We generated nine new rat monoclonal antibodies specific to mouse Siglec-E, with no cross-reactivity to Siglec-F. Although all antibodies detected Siglec-E on transfected human HEK-293T cells, only two reacted with mouse bone marrow neutrophils by flow cytometry and on spleen sections by immunohistochemistry. Moreover, whereas all antibodies recognized Siglec-E-Fc on immunoblots, binding was dependent on intact disulfide bonds and N-glycans, and only two antibodies recognized native Siglec-E within spleen lysates. Thus, we further investigated the impact of Siglec-E homodimerization. Homology-based structural modeling predicted a cysteine residue (Cys-298) in position to form a disulfide bridge between two Siglec-E polypeptides. Mutagenesis of Cys-298 confirmed its role in dimerization. In keeping with the high level of 9-O-acetylation found in mice, sialoglycan array studies indicate that this modification has complex effects on recognition by Siglec-E, in relationship to the underlying structures. However, we found no differences in phosphorylation or SHP-1 recruitment between dimeric and monomeric Siglec-E expressed on HEK293A cells. Phylogenomic analyses predicted that only some human and mouse Siglecs form disulfide-linked dimers. Notably, Siglec-9, the functionally equivalent human paralog of Siglec-E, occurs as a monomer.


Sujet(s)
Antigènes CD/métabolisme , Antigènes de différenciation des lymphocytes B/métabolisme , Régulation de l'expression des gènes/physiologie , Multimérisation de protéines/physiologie , Substitution d'acide aminé , Animaux , Anticorps/composition chimique , Antigènes CD/composition chimique , Antigènes CD/génétique , Antigènes de différenciation des lymphocytes B/composition chimique , Antigènes de différenciation des lymphocytes B/génétique , Cellules dendritiques/cytologie , Cellules dendritiques/métabolisme , Glycosylation , Humains , Macrophages/cytologie , Macrophages/métabolisme , Souris , Souris knockout , Monocytes/cytologie , Monocytes/métabolisme , Mutagenèse , Mutation faux-sens , Granulocytes neutrophiles/cytologie , Granulocytes neutrophiles/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Rats , Rats de lignée LEW , Lectines liant l'acide sialique apparentées aux immunoglobulines/composition chimique , Lectines liant l'acide sialique apparentées aux immunoglobulines/génétique , Lectines liant l'acide sialique apparentées aux immunoglobulines/métabolisme
12.
Front Immunol ; 8: 1926, 2017.
Article de Anglais | MEDLINE | ID: mdl-29379501

RÉSUMÉ

Siglec-E is a murine CD33-related siglec that functions as an inhibitory receptor and is expressed mainly on neutrophils and macrophage populations. Recent studies have suggested that siglec-E is an important negative regulator of lipopolysaccharide (LPS)-toll-like receptor 4 (TLR4) signaling and one report (1) claimed that siglec-E is required for TLR4 endocytosis following uptake of Escherichia coli by macrophages and dendritic cells (DCs). Our attempts to reproduce these observations using cells from wild-type (WT) and siglec-E-deficient mice were unsuccessful. We used a variety of assays to determine if siglec-E expressed by different macrophage populations can regulate TLR4 signaling in response to LPS, but found no consistent differences in cytokine secretion in vitro and in vivo, comparing three different strains of siglec-E-deficient mice with matched WT controls. No evidence was found that the siglec-E deficiency was compensated by expression of siglecs-F and -G, the other murine inhibitory CD33-related siglecs. Quantitative proteomics was used as an unbiased approach and provided additional evidence that siglec-E does not suppress inflammatory TLR4 signaling. Interestingly, proteomics revealed a siglec-E-dependent alteration in macrophage protein composition that could be relevant to functional responses in host defense. In support of this, siglec-E-deficient mice exhibited enhanced growth of Salmonella enterica serovar Typhimurium in the liver following intravenous infection, but macrophages lacking siglec-E did not show altered uptake or killing of bacteria in vitro. Using various cell types including bone marrow-derived DCs (BMDCs), splenic DCs, and macrophages from WT and siglec-E-deficient mice, we showed that siglec-E is not required for TLR4 endocytosis following E. coli uptake or LPS challenge. We failed to see expression of siglec-E by BMDC even after LPS-induced maturation, but confirmed previous studies that splenic DCs express low levels of siglec-E. Taken together, our findings do not support a major role of siglec-E in regulation of TLR4 signaling functions or TLR4 endocytosis in macrophages or DCs. Instead, they reveal that induction of siglec-E by LPS can modulate the phenotype of macrophages, the functional significance of which is currently unclear.

13.
J Biol Chem ; 289(29): 20370-6, 2014 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-24895121

RÉSUMÉ

Siglec-E is a sialic acid-binding Ig-like lectin expressed on murine myeloid cells. It has recently been shown to function as a negative regulator of ß2-integrin-dependent neutrophil recruitment to the lung following exposure to lipopolysaccharide (LPS). Here, we demonstrate that siglec-E promoted neutrophil production of reactive oxygen species (ROS) following CD11b ß2-integrin ligation with fibrinogen in a sialic acid-dependent manner, but it had no effect on ROS triggered by a variety of other stimulants. Siglec-E promotion of ROS was likely mediated via Akt activation, because siglec-E-deficient neutrophils plated on fibrinogen exhibited reduced phosphorylation of Akt, and the Akt inhibitor, MK2206, blocked fibrinogen-induced ROS. In vivo imaging showed that siglec-E also promoted ROS in acutely inflamed lungs following exposure of mice to LPS. Importantly, siglec-E-promoted ROS were required for its inhibitory function, as the NADPH oxidase inhibitor, apocynin, reversed the siglec-E-mediated suppression of neutrophil recruitment and blocked neutrophil ROS production in vitro. Taken together, these results demonstrate that siglec-E functions as an inhibitory receptor of neutrophils via positive regulation of NADPH oxidase activation and ROS production. Our findings have implications for the inhibitory role of siglec-9 on human neutrophils in sepsis and acute lung injury.


Sujet(s)
Antigènes CD/métabolisme , Antigènes de différenciation des lymphocytes B/métabolisme , Antigènes CD18/métabolisme , Poumon/immunologie , Poumon/métabolisme , NADPH oxidase/métabolisme , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Lésion pulmonaire aigüe/immunologie , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/anatomopathologie , Substitution d'acide aminé , Animaux , Antigènes CD/composition chimique , Antigènes CD/génétique , Antigènes de différenciation des lymphocytes B/composition chimique , Antigènes de différenciation des lymphocytes B/génétique , Mouvement cellulaire , Activation enzymatique , Femelle , Fibrinogène/métabolisme , Composés hétérocycliques 3 noyaux/pharmacologie , Humains , Lipopolysaccharides/toxicité , Poumon/anatomopathologie , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Souches mutantes de souris , Mutagenèse dirigée , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes c-akt/antagonistes et inhibiteurs , Protéines proto-oncogènes c-akt/métabolisme , Espèces réactives de l'oxygène/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE