Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 132
Filtrer
1.
Front Immunol ; 15: 1422370, 2024.
Article de Anglais | MEDLINE | ID: mdl-38938578

RÉSUMÉ

Introduction: Hematopoietic stem cell transplantation (HSCT) is associated with immune complications and endothelial dysfunction due to intricate donor-recipient interactions, conditioning regimens, and inflammatory responses. Methods: This study investigated the role of the complement system during HSCT and its interaction with the cytokine network. Seventeen acute myeloid leukemia patients undergoing HSCT were monitored, including blood sampling from the start of the conditioning regimen until four weeks post-transplant. Clinical follow-up was 200 days. Results: Total complement functional activity was measured by WIELISA and the degree of complement activation by ELISA measurement of sC5b-9. Cytokine release was measured using a 27-multiplex immuno-assay. At all time-points during HSCT complement functional activity remained comparable to healthy controls. Complement activation was continuously stable except for two patients demonstrating increased activation, consistent with severe endotheliopathy and infections. In vitro experiments with post-HSCT whole blood challenged with Escherichia coli, revealed a hyperinflammatory cytokine response with increased TNF, IL-1ß, IL-6 and IL-8 formation. Complement C3 inhibition markedly reduced the cytokine response induced by Staphylococcus aureus, Aspergillus fumigatus, and cholesterol crystals. Discussion: In conclusion, HSCT patients generally retained a fully functional complement system, whereas activation occurred in patients with severe complications. The complement-cytokine interaction indicates the potential for new complement-targeting therapeutic strategies in HSCT.


Sujet(s)
Activation du complément , Cytokines , Transplantation de cellules souches hématopoïétiques , Transplantation homologue , Humains , Mâle , Transplantation de cellules souches hématopoïétiques/effets indésirables , Femelle , Adulte d'âge moyen , Adulte , Cytokines/métabolisme , Sujet âgé , Leucémie aigüe myéloïde/thérapie , Leucémie aigüe myéloïde/immunologie , Protéines du système du complément/immunologie , Protéines du système du complément/métabolisme , Conditionnement pour greffe/méthodes , Jeune adulte
2.
Front Immunol ; 15: 1383505, 2024.
Article de Anglais | MEDLINE | ID: mdl-38686379

RÉSUMÉ

Acute myocardial infarction (MI) results in tissue damage to affected areas of the myocardium. The initial inflammatory response is the most damaging for residual cardiac function, while at later stages inflammation is a prerequisite for proper healing and scar formation. Balancing the extent and duration of inflammation during various stages after MI is thus pivotal for preserving cardiac function. Recently, a signaling lymphocytic activation molecule 1 (SLAMF1)-derived peptide (P7) was shown to reduce the secretion of inflammatory cytokines and protected against acute lipopolysaccharide-induced death in mice. In the present study, we experimentally induced MI by permanent ligation of the left anterior descending artery (LAD) in mice and explored the beneficial effect of immediately administering P7, with the aim of dampening the initial inflammatory phase without compromising the healing and remodeling phase. Blood samples taken 9 h post-LAD surgery and P7 administration dampened the secretion of inflammatory cytokines, but this dampening effect of P7 was diminished after 3 days. Echocardiography revealed less deterioration of cardiac contraction in mice receiving P7. In line with this, less myocardial damage was observed histologically in P7-treated mice. In conclusion, the administration of a SLAMF1-derived peptide (P7) immediately after induction of MI reduces the initial myocardial inflammation, reduces infarct expansion, and leads to less deterioration of cardiac contraction.


Sujet(s)
Modèles animaux de maladie humaine , Infarctus du myocarde , Animaux , Souris , Mâle , Cytokines/métabolisme , Souris de lignée C57BL , Antigènes CD/métabolisme , Ligature , Myocarde/anatomopathologie , Myocarde/métabolisme , Peptides/pharmacologie , Récepteurs de surface cellulaire/métabolisme , Vaisseaux coronaires/effets des médicaments et des substances chimiques , Vaisseaux coronaires/anatomopathologie
3.
Life Sci Alliance ; 6(12)2023 12.
Article de Anglais | MEDLINE | ID: mdl-37788908

RÉSUMÉ

Inflammation plays a crucial role in the development and progression of many diseases, and is often caused by dysregulation of signalling from pattern recognition receptors, such as TLRs. Inhibition of key protein-protein interactions is an attractive target for treating inflammation. Recently, we demonstrated that the signalling lymphocyte activation molecule family 1 (SLAMF1) positively regulates signalling downstream of TLR4 and identified the interaction interface between SLAMF1 and the TLR4 adaptor protein TRIF-related adapter molecule (TRAM). Based on these findings, we developed a SLAMF1-derived peptide, P7, which is linked to a cell-penetrating peptide for intracellular delivery. We found that P7 peptide inhibits the expression and secretion of IFNß and pro-inflammatory cytokines (TNF, IL-1ß, IL-6) induced by TLR4, and prevents death in mice subjected to LPS shock. The mechanism of action of P7 peptide is based on interference with several intracellular protein-protein interactions, including TRAM-SLAMF1, TRAM-Rab11FIP2, and TIRAP-MyD88 interactions. Overall, P7 peptide has a unique mode of action and demonstrates high efficacy in inhibiting TLR4-mediated signalling in vitro and in vivo.


Sujet(s)
Transduction du signal , Récepteur de type Toll-4 , Animaux , Souris , Famille des molécules de signalisation de l'activation des lymphocytes/métabolisme , Peptides/pharmacologie , Protéines adaptatrices de la transduction du signal/métabolisme , Inflammation
4.
Cell Rep Med ; 4(7): 101098, 2023 07 18.
Article de Anglais | MEDLINE | ID: mdl-37390829

RÉSUMÉ

During differentiation, neutrophils undergo a spontaneous pro-inflammatory program that is hypothesized here to be under caspase-8 control. In mice, intraperitoneal administration of the caspase-8 inhibitor z-IETD-fmk is sufficient to unleash the production of pro-inflammatory cytokines and neutrophil influx in the absence of cell death. These effects are due to selective inhibition of caspase-8 and require tonic interferon-ß (IFN-ß) production and RIPK3 but not MLKL, the essential downstream executioner of necroptotic cell death. In vitro, stimulation with z-IETD-fmk is sufficient to induce significant cytokine production in murine neutrophils but not in macrophages. Therapeutic administration of z-IETD-fmk improves clinical outcome in models of lethal bacterial peritonitis and pneumonia by augmenting cytokine release, neutrophil influx, and bacterial clearance. Moreover, the inhibitor protects mice against high-dose endotoxin shock. Collectively, our data unveil a RIPK3- and IFN-ß-dependent pathway that is constitutively activated in neutrophils and can be harnessed therapeutically using caspase-8 inhibition.


Sujet(s)
Apoptose , Infections bactériennes , Animaux , Souris , Infections bactériennes/traitement médicamenteux , Caspase 8/métabolisme , Caspase 8/pharmacologie , Cytokines/métabolisme , Activation des neutrophiles
5.
Pediatr Res ; 94(2): 512-519, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-36725909

RÉSUMÉ

BACKGROUND: Escherichia coli and Group B streptococci (GBS) are the main causes of neonatal early-onset sepsis (EOS). Despite antibiotic therapy, EOS is associated with high morbidity and mortality. Dual inhibition of complement C5 and the Toll-like receptor co-factor CD14 has in animal studies been a promising novel therapy for sepsis. METHODS: Whole blood was collected from the umbilical cord after caesarean section (n = 30). Blood was anti-coagulated with lepirudin. C5 inhibitor (eculizumab) and anti-CD14 was added 8 min prior to, or 15 and 30 min after adding E. coli or GBS. Total bacterial incubation time was 120 min (n = 16) and 240 min (n = 14). Cytokines and the terminal complement complex (TCC) were measured using multiplex technology and ELISA. RESULTS: Dual inhibition significantly attenuated TCC formation by 25-79% when adding inhibitors with up to 30 min delay in both E. coli- and GBS-induced inflammation. TNF, IL-6 and IL-8 plasma concentration were significantly reduced by 28-87% in E. coli-induced inflammation when adding inhibitors with up to 30 min delay. The dual inhibition did not significantly reduce TNF, IL-6 and IL-8 plasma concentration in GBS-induced inflammation. CONCLUSION: Dual inhibition of C5 and CD14 holds promise as a potential future treatment for severe neonatal EOS. IMPACT: Neonatal sepsis can cause severe host inflammation with high morbidity and mortality, but there are still no effective adjunctive immunologic interventions available. Adding CD14 and complement C5 inhibitors up to 30 min after incubation of E. coli or Group B streptococci in a human umbilical cord blood model significantly reduced complement activation and cytokine release. Dual inhibition of C5 and CD14 is a potential future therapy to modulate systemic inflammation in severe cases of neonatal sepsis.


Sujet(s)
Sepsis néonatal , Sepsie , Grossesse , Animaux , Nouveau-né , Humains , Femelle , Complément C5 , Escherichia coli , Sang foetal , Interleukine-6 , Interleukine-8 , Césarienne , Cytokines , Inflammation , Antigènes CD14
6.
iScience ; 26(1): 105895, 2023 Jan 20.
Article de Anglais | MEDLINE | ID: mdl-36590899

RÉSUMÉ

COVID-19 pandemic continues to remain a global health concern owing to the emergence of newer variants. Several multi-Omics studies have produced extensive evidence on host-pathogen interactions and potential therapeutic targets. Nonetheless, an increased understanding of host signaling networks regulated by post-translational modifications and their ensuing effect on the cellular dynamics is critical to expanding the current knowledge on SARS-CoV-2 infections. Through an unbiased transcriptomics, proteomics, acetylomics, phosphoproteomics, and exometabolome analysis of a lung-derived human cell line, we show that SARS-CoV-2 Norway/Trondheim-S15 strain induces time-dependent alterations in the induction of type I IFN response, activation of DNA damage response, dysregulated Hippo signaling, among others. We identified interplay of phosphorylation and acetylation dynamics on host proteins and its effect on the altered release of metabolites, especially organic acids and ketone bodies. Together, our findings serve as a resource of potential targets that can aid in designing novel host-directed therapeutic strategies.

7.
Biomedicines ; 10(7)2022 Jun 22.
Article de Anglais | MEDLINE | ID: mdl-35884781

RÉSUMÉ

Toll-like receptor 8 (TLR8) recognizes single-stranded RNA of viral and bacterial origin as well as mediates the secretion of pro-inflammatory cytokines and type I interferons by human monocytes and macrophages. TLR8, as other endosomal TLRs, utilizes the MyD88 adaptor protein for initiation of signaling from endosomes. Here, we addressed the potential role of the Toll-interleukin 1 receptor domain-containing adaptor protein (TIRAP) in the regulation of TLR8 signaling in human primary monocyte-derived macrophages (MDMs). To accomplish this, we performed TIRAP gene silencing, followed by the stimulation of cells with synthetic ligands or live bacteria. Cytokine-gene expression and secretion were analyzed by quantitative PCR or Bioplex assays, respectively, while nuclear translocation of transcription factors was addressed by immunofluorescence and imaging, as well as by cell fractionation and immunoblotting. Immunoprecipitation and Akt inhibitors were also used to dissect the signaling mechanisms. Overall, we show that TIRAP is recruited to the TLR8 Myddosome signaling complex, where TIRAP contributes to Akt-kinase activation and the nuclear translocation of interferon regulatory factor 5 (IRF5). Recruitment of TIRAP to the TLR8 signaling complex promotes the expression and secretion of the IRF5-dependent cytokines IFNß and IL-12p70 as well as, to a lesser degree, TNF. These findings reveal a new and unconventional role of TIRAP in innate immune defense.

8.
J Immunol ; 208(12): 2771-2778, 2022 06 15.
Article de Anglais | MEDLINE | ID: mdl-35675954

RÉSUMÉ

Thrombin plays a central role in thromboinflammatory responses, but its activity is blocked in the common ex vivo human whole blood models, making an ex vivo study of thrombin effects on thromboinflammatory responses unfeasible. In this study, we exploited the anticoagulant peptide Gly-Pro-Arg-Pro (GPRP) that blocks fibrin polymerization to study the effects of thrombin on acute inflammation in response to Escherichia coli and Staphylococcus aureus Human blood was anticoagulated with either GPRP or the thrombin inhibitor lepirudin and incubated with either E. coli or S. aureus for up to 4 h at 37°C. In GPRP-anticoagulated blood, there were spontaneous elevations in thrombin levels and platelet activation, which further increased in the presence of bacteria. Complement activation and the expression of activation markers on monocytes and granulocytes increased to the same extent in both blood models in response to bacteria. Most cytokines were not elevated in response to thrombin alone, but thrombin presence substantially and heterogeneously modulated several cytokines that increased in response to bacterial incubations. Bacterial-induced releases of IL-8, MIP-1α, and MIP-1ß were potentiated in the thrombin-active GPRP model, whereas the levels of IP-10, TNF, IL-6, and IL-1ß were elevated in the thrombin-inactive lepirudin model. Complement C5-blockade, combined with CD14 inhibition, reduced the overall cytokine release significantly, both in thrombin-active and thrombin-inactive models. Our data support that thrombin itself marginally induces leukocyte-dependent cytokine release in this isolated human whole blood but is a significant modulator of bacteria-induced inflammation by a differential effect on cytokine patterns.


Sujet(s)
Infections à Escherichia coli , Infections à staphylocoques , Cytokines/métabolisme , Escherichia coli/physiologie , Humains , Inflammation , Staphylococcus aureus/métabolisme , Thrombine/métabolisme
9.
Mol Divers ; 26(4): 2175-2188, 2022 Aug.
Article de Anglais | MEDLINE | ID: mdl-34668104

RÉSUMÉ

IFN-ß is a cytokine that plays a significant role in the immune system. Inhibition of IFN-ß might be used as a therapeutic approach to treat septic shock. A peptidomimetic previously developed by our research team, 1-benzyl-5-methyl-4-(n-octylamino)pyrimidin-2(1H)-one (LT87), was used as an cardioprotective agent in a myocardial ischemia (MI) mouse model. We have developed new LT87 derivatives by synthetizing its dimers in an attempt to extend its structural variety and enhance its biological activity. A dimeric derivative, LT127, exhibited a dose-dependent inhibition of LPS-mediated IFN-ß and subsequent CXCL10 mRNA transcription. The effect was selective and transduced through TLR4- and TRAM/TRIF-mediated signaling, with no significant effect on MyD88-dependent signaling. However, this effect was not specific to TLR4, since a similar effect was observed both on TLR8- and MDA5/RIG-I-stimulated IFN-ß expression. Nevertheless, LT127 might serve as a drug candidate, specifically as an inhibitor for IFN-ß production in order to develop a novel therapeutic approach to prevent septic shock.


Sujet(s)
Interféron bêta , Peptidomimétiques , Choc septique , Animaux , Cytokines/métabolisme , Interféron bêta/métabolisme , Souris , Peptidomimétiques/pharmacologie , Choc septique/traitement médicamenteux , Transduction du signal , Récepteur de type Toll-4/génétique , Récepteur de type Toll-4/métabolisme
10.
Sci Immunol ; 6(66): eabf2489, 2021 Dec 24.
Article de Anglais | MEDLINE | ID: mdl-34932384

RÉSUMÉ

While serum-circulating complement destroys invading pathogens, intracellularly active complement, termed the "complosome," functions as a vital orchestrator of cell-metabolic events underlying T cell effector responses. Whether intracellular complement is also nonredundant for the activity of myeloid immune cells is currently unknown. Here, we show that monocytes and macrophages constitutively express complement component (C) 5 and generate autocrine C5a via formation of an intracellular C5 convertase. Cholesterol crystal sensing by macrophages induced C5aR1 signaling on mitochondrial membranes, which shifted ATP production via reverse electron chain flux toward reactive oxygen species generation and anaerobic glycolysis to favor IL-1ß production, both at the transcriptional level and processing of pro­IL-1ß. Consequently, atherosclerosis-prone mice lacking macrophage-specific C5ar1 had ameliorated cardiovascular disease on a high-cholesterol diet. Conversely, inflammatory gene signatures and IL-1ß produced by cells in unstable atherosclerotic plaques of patients were normalized by a specific cell-permeable C5aR1 antagonist. Deficiency of the macrophage cell-autonomous C5 system also protected mice from crystal nephropathy mediated by folic acid. These data demonstrate the unexpected intracellular formation of a C5 convertase and identify C5aR1 as a direct modulator of mitochondrial function and inflammatory output from myeloid cells. Together, these findings suggest that the complosome is a contributor to the biologic processes underlying sterile inflammation and indicate that targeting this system could be beneficial in macrophage-dependent diseases, such as atherosclerosis.


Sujet(s)
Inflammation/immunologie , Interleukine-1 bêta/biosynthèse , Macrophages/immunologie , Récepteur à l'anaphylatoxine C5a/immunologie , Animaux , Lignée cellulaire , Humains , Souris , Souris de lignée C57BL , Souris knockout , Récepteur à l'anaphylatoxine C5a/déficit
11.
PLoS One ; 16(10): e0258989, 2021.
Article de Anglais | MEDLINE | ID: mdl-34705862

RÉSUMÉ

Toll-like receptors (TLRs) are highly-conserved pattern recognition receptors that mediate innate immune responses to invading pathogens and endogenous danger signals released from damaged and dying cells. Activation of TLRs trigger downstream signaling cascades, that culminate in the activation of interferon regulatory factors (IRFs), which subsequently leads to type I interferon (IFN) response. In the current study, we sought to expand the scope of gene expression changes in THP1-derived macrophages upon TLR4 activation and to identify interferon-stimulated genes. RNA-seq analysis led to the identification of several known and novel differentially expressed genes, including CMPK2, particularly in association with type I IFN signaling. We performed an in-depth characterization of CMPK2 expression, a nucleoside monophosphate kinase that supplies intracellular UTP/CTP for nucleic acid synthesis in response to type I IFN signaling in macrophages. CMPK2 was significantly induced at both RNA and protein levels upon stimulation with TLR4 ligand-LPS and TLR3 ligand-Poly (I:C). Confocal microscopy and subcellular fractionation indicated CMPK2 localization in both cytoplasm and mitochondria of THP-1 macrophages. Furthermore, neutralizing antibody-based inhibition of IFNAR receptor in THP-1 cells and BMDMs derived from IFNAR KO and IRF3 KO knockout mice further revealed that CMPK2 expression is dependent on LPS/Poly (I:C) mediated IRF3- type I interferon signaling. In summary, our findings suggest that CMPK2 is a potential interferon-stimulated gene in THP-1 macrophages and that CMPK2 may facilitate IRF3- type I IFN-dependent anti-bacterial and anti-viral roles.


Sujet(s)
Expression des gènes/immunologie , Facteur-3 de régulation d'interféron/immunologie , Macrophages/métabolisme , Nucleoside phosphate kinase/immunologie , Récepteur à l'interféron alpha-bêta/immunologie , Animaux , Humains , Macrophages/cytologie , Souris , Souris knockout , Cellules THP-1
12.
Acta Biomater ; 127: 169-179, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33785451

RÉSUMÉ

Inadequate supplies of donor corneas have evoked an escalating interest in corneal xenotransplantation. However, innate immune responses contribute significantly to the mechanism of xenograft rejection. We hypothesized that complement component C5 and TLR co-receptor CD14 inhibition would inhibit porcine cornea induced innate immune responses. Therefore, we measured cytokine release in human blood, induced by three forms of corneal xenografts with or without inhibitors. Native porcine cornea (NPC) induced interleukins (IL-1ß, IL-2, IL-6, IL-8, IL-1ra), chemokines (MCP-1, MIP-1α, MIP-1ß) and other cytokines (TNF, G-CSF, INF-γ, FGF-basic). Decellularized (DPC) and gamma-irradiated cornea (g-DPC) elevated the release of those cytokines. C5-blockade by eculizumab inhibited all the cytokines except G-CSF when induced by NPC. However, C5-blockade failed to reduce DPC and g-DPC induced cytokines. Blockade of CD14 inhibited DPC-induced cytokines except for IL-8, MCP-1, MIP-1α, and G-CSF, while it inhibited all of them when induced by g-DPC. Combined blockade of C5 and CD14 inhibited the maximum number of cytokines regardless of the xenograft type. Finally, by using the TLR4 specific inhibitor Eritoran, we showed that TLR4 activation was the basis for the CD14 effect. Thus, blockade of C5, when combined with TLR4 inhibition, may have therapeutic potential in pig-to-human corneal xenotransplantation. STATEMENT OF SIGNIFICANCE: Bio-engineered corneal xenografts are on the verge of becoming a viable alternative to allogenic human-donor-cornea, but the host's innate immune response is still a critical barrier for graft acceptance. By overruling this barrier, limited graft availability would no longer be an issue for treating corneal diseases. We showed that the xenograft induced inflammation is initiated by the complement system and toll-like receptor activation. Intriguingly, the inflammatory response was efficiently blocked by simultaneously targeting bottleneck molecules in the complement system (C5) and the TLR co-receptor CD14 with pharmaceutical inhibitors. We postulate that a combination of C5 and CD14 inhibition could have a great therapeutic potential to overcome the immunologic barrier in pig-to-human corneal xenotransplantation.


Sujet(s)
Complément C5/antagonistes et inhibiteurs , Transplantation de cornée/effets indésirables , Hétérogreffes , Inflammation/étiologie , Antigènes CD14 , Animaux , Cornée , Cytokines , Humains , Suidae , Transplantation hétérologue
13.
EBioMedicine ; 60: 102985, 2020 Oct.
Article de Anglais | MEDLINE | ID: mdl-32927275

RÉSUMÉ

BACKGROUND: During atherogenesis, cholesterol precipitates into cholesterol crystals (CC) in the vessel wall, which trigger plaque inflammation by activating the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome. We investigated the relationship between CC, complement and NLRP3 in patients with cardiovascular disease. METHODS: We analysed plasma, peripheral blood mononuclear cells (PBMC) and carotid plaques from patients with advanced atherosclerosis applying ELISAs, multiplex cytokine assay, qPCR, immunohistochemistry, and gene profiling. FINDINGS: Transcripts of interleukin (IL)-1beta(ß) and NLRP3 were increased and correlated in PBMC from patients with acute coronary syndrome (ACS). Priming of these cells with complement factor 5a (C5a) and tumour necrosis factor (TNF) before incubation with CC resulted in increased IL-1ß protein when compared to healthy controls. As opposed to healthy controls, systemic complement was significantly increased in patients with stable angina pectoris or ACS. In carotid plaques, complement C1q and C5b-9 complex accumulated around CC-clefts, and complement receptors C5aR1, C5aR2 and C3aR1 were higher in carotid plaques compared to control arteries. Priming human carotid plaques with C5a followed by CC incubation resulted in pronounced release of IL-1ß, IL-18 and IL-1α. Additionally, mRNA profiling demonstrated that C5a and TNF priming followed by CC incubation upregulated plaque expression of NLRP3 inflammasome components. INTERPRETATION: We demonstrate that CC are important local- and systemic complement activators, and we reveal that the interaction between CC and complement could exert its effect by activating the NLRP3 inflammasome, thus promoting the progression of atherosclerosis.


Sujet(s)
Artériopathies carotidiennes/étiologie , Artériopathies carotidiennes/métabolisme , Cholestérol/métabolisme , Protéines du système du complément/immunologie , Maladie des artères coronaires/étiologie , Maladie des artères coronaires/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Transduction du signal , Artériopathies carotidiennes/anatomopathologie , Complément C5a/immunologie , Biologie informatique/méthodes , Maladie des artères coronaires/anatomopathologie , Cytokines/métabolisme , Prédisposition aux maladies , Analyse de profil d'expression de gènes , Humains , Inflammasomes/métabolisme , Médiateurs de l'inflammation/métabolisme , Agranulocytes/immunologie , Agranulocytes/métabolisme , Agranulocytes/anatomopathologie , Cristaux liquides , Plaque d'athérosclérose
14.
J Leukoc Biol ; 107(4): 673-683, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-32083344

RÉSUMÉ

We recently showed that TLR8 is critical for the detection of Gram-positive bacteria by human monocytes. Here, we hypothesized that TLR8 and complement together regulate antibacterial responses in human blood. Anticoagulated blood was treated with selective inhibitors of TLR8 and/or complement C5, and then challenged with live Streptococcus agalactiae (Group B streptococcus, GBS), Staphylococcus aureus, or Escherichia coli. Cytokine production, plasma membrane permeability, bacterial survival, phagocytosis, and activation of coagulation was examined. GBS and S. aureus, but not E. coli, triggered TLR8-dependent production of IL-12p70, IL-1ß, TNF, and IL-6 in fresh human whole blood. In purified polymorphonuclear neutrophils (PMN), GBS and S. aureus induced IL-8 release in part via TLR8, whereas PMN plasma membrane leakage and extracellular DNA levels increased independently of TLR8. TLR8 was more important than C5 for bacteria-induced production of IL-12p70, IL-1ß, and TNF in blood, whereas IL-8 release was more C5 dependent. Both TLR8 and C5 induced IL-6 release and activation of prothrombin cleavage, and here their combined effects were additive. Blocking of C5 or C5aR1 attenuated phagocytosis and increased the extracellular growth of GBS in blood, whereas TLR8 inhibition neither reduced phagocytosis nor intracellular killing of GBS and S. aureus. In conclusion, TLR8 is more important than C5 for production of IL-12p70, IL-1ß, and TNF upon GBS and S. aureus infection in blood, whereas C5 is central for IL-8 release and phagocytosis. Both TLR8 and C5 mediate IL-6 release and activation of coagulation during challenge with Gram-positive bacteria in blood.


Sujet(s)
Complément C5/métabolisme , Cytokines/sang , Bactéries à Gram positif/physiologie , Thrombine/métabolisme , Récepteur de type Toll-8/sang , Coagulation sanguine , Membrane cellulaire/métabolisme , Survie cellulaire , ADN/métabolisme , Humains , Interleukine-8/métabolisme , Antigènes CD14/métabolisme , Viabilité microbienne , Monocytes/métabolisme , Granulocytes neutrophiles/métabolisme , Récepteur de type Toll-8/antagonistes et inhibiteurs , Récepteur de type Toll-8/métabolisme
15.
Atherosclerosis ; 296: 74-82, 2020 03.
Article de Anglais | MEDLINE | ID: mdl-31870625

RÉSUMÉ

BACKGROUND AND AIMS: We have previously found increased levels of the cysteine protease legumain in plasma and plaques from patients with carotid atherosclerosis. This study further investigated legumain during acute cardiovascular events. METHODS: Circulating levels of legumain from patients and legumain released from platelets were assessed by enzyme-linked-immunosorbent assay. Quantitative PCR and immunoblotting were used to study expression, while localization was visualized by immunohistochemistry. RESULTS: In the SUMMIT Malmö cohort (n = 339 with or without type 2 diabetes and/or cardiovascular disease [CVD], and 64 healthy controls), the levels of circulating legumain were associated with the presence of CVD in non-diabetics, with no relation to outcome. In symptomatic carotid plaques and in samples from both coronary and intracerebral thrombi obtained during acute cardiovascular events, legumain was co-localized with macrophages in the same regions as platelets. In vitro, legumain was shown to be present in and released from platelets upon activation. In addition, THP-1 macrophages exposed to releasate from activated platelets showed increased legumain expression. Interestingly, primary peripheral blood mononuclear cells stimulated with recombinant legumain promoted anti-inflammatory responses. Finally, in a STEMI population (POSTEMI; n = 272), patients had significantly higher circulating legumain before and immediately after percutaneous coronary intervention compared with healthy controls (n = 67), and high levels were associated with improved outcome. CONCLUSIONS: Our data demonstrate for the first time that legumain is upregulated during acute cardiovascular events and is associated with improved outcome.


Sujet(s)
Maladies cardiovasculaires/métabolisme , Cysteine endopeptidases/biosynthèse , Macrophages/enzymologie , Infarctus du myocarde avec sus-décalage du segment ST/sang , Maladie aigüe , Séquence d'acides aminés , Plaquettes/métabolisme , Maladies cardiovasculaires/complications , Maladies cardiovasculaires/anatomopathologie , Artériopathies carotidiennes/métabolisme , Artériopathies carotidiennes/anatomopathologie , Études transversales , Cysteine endopeptidases/sang , Cysteine endopeptidases/génétique , Cysteine endopeptidases/pharmacologie , Cytokines/pharmacologie , Diabète de type 2/sang , Diabète de type 2/complications , Études de suivi , Humains , Lipopolysaccharides/pharmacologie , Monocytes/effets des médicaments et des substances chimiques , Intervention coronarienne percutanée , Plaque d'athérosclérose/composition chimique , Activation plaquettaire , Protéines recombinantes/pharmacologie , Infarctus du myocarde avec sus-décalage du segment ST/mortalité , Infarctus du myocarde avec sus-décalage du segment ST/chirurgie , Suède/épidémiologie , Cellules THP-1
16.
MAbs ; 12(1): 1686319, 2020.
Article de Anglais | MEDLINE | ID: mdl-31671278

RÉSUMÉ

The mechanism of action of recombinant IgG2/4 antibodies involves blocking of their target without the induction of effector functions. Examples are eculizumab (Soliris®), which is used clinically to block complement factor C5, as well as anti-human CD14 (r18D11) and anti-porcine CD14 (rMIL2) produced in our laboratory. So far, no proper IgG2/4 control antibody has been available for controlled validation of IgG2/4 antibody functions. Here, we describe the design of a recombinant control antibody (NHDL), which was generated by combining the variable light (VL) and heavy (VH) chains from two unrelated specificities. NHDL was readily expressed and purified as a stable IgG2/4 antibody, and showed no detectable specificity toward any putative antigen present in human or porcine blood. The approach of artificial VL/VH combination may be adopted for the design of other recombinant control antibodies.


Sujet(s)
Anticorps monoclonaux/génétique , Immunoglobuline G/génétique , Chaines lourdes des immunoglobulines/génétique , Chaines légères des immunoglobulines/génétique , Protéines de fusion recombinantes/génétique , Animaux , Anticorps monoclonaux/métabolisme , Biothérapie , Protéines du sang/métabolisme , Épitopes/métabolisme , Humains , Antigènes CD14/immunologie , Antigènes CD14/métabolisme , Souris , Placebo , Ingénierie des protéines , Suidae
17.
J Immunol ; 203(6): 1571-1578, 2019 09 15.
Article de Anglais | MEDLINE | ID: mdl-31413105

RÉSUMÉ

Heme is a critical danger molecule liberated from hemeproteins in various conditions, including from hemoglobin in hemolytic diseases. Heme may cause thromboinflammatory damage by activating inflammatory and hemostatic pathways, such as complement, the TLRs, coagulation, and platelets. In this study, we explored the effect of single and dual inhibition of complement component C5 and TLR coreceptor CD14 on heme-induced thromboinflammation in an ex vivo human whole blood model. Heme induced a dose-dependent activation of complement via the alternative pathway. Single inhibition of C5 by eculizumab attenuated the release of IL-6, IL-8, TNF, MCP-1, MIP-1α, IFN-γ, LTB-4, MMP-8 and -9, and IL-1Ra with more than 60% (p < 0.05 for all) reduced the upregulation of CD11b on granulocytes and monocytes by 59 and 40%, respectively (p < 0.05), and attenuated monocytic tissue factor expression by 33% (p < 0.001). Blocking CD14 attenuated IL-6 and TNF by more than 50% (p < 0.05). In contrast to single inhibition, combined C5 and CD14 was required for a significantly attenuated prothrombin cleavage (72%, p < 0.05). Markers of thromboinflammation were also quantified in two patients admitted to the hospital with sickle cell disease (SCD) crisis. Both SCD patients had pronounced hemolysis and depleted plasma hemopexin and haptoglobin. Plasma heme and complement activation was markedly increased in one patient, a coinciding observation as demonstrated ex vivo. In conclusion, heme-induced thromboinflammation was largely attenuated by C5 inhibition alone, with a beneficial effect of adding a CD14 inhibitor to attenuate prothrombin activation. Targeting C5 has the potential to reduce thromboinflammation in SCD crisis patients.


Sujet(s)
Complément C5/métabolisme , Hème/métabolisme , Inflammation/métabolisme , Antigènes CD14/métabolisme , Adulte , Drépanocytose/métabolisme , Animaux , Coagulation sanguine/physiologie , Activation du complément/physiologie , Cytokines/métabolisme , Granulocytes/métabolisme , Hémolyse/physiologie , Humains , Mâle , Monocytes/métabolisme , Suidae , Thromboplastine/métabolisme
18.
J Immunol ; 203(4): 853-863, 2019 08 15.
Article de Anglais | MEDLINE | ID: mdl-31270150

RÉSUMÉ

Cholesterol crystals (CC) are strong activators of complement and could potentially be involved in thromboinflammation through complement-coagulation cross-talk. To explore the coagulation-inducing potential of CC, we performed studies in lepirudin-based human whole blood and plasma models. In addition, immunohistological examinations of brain thrombi and vulnerable plaque material from patients with advanced carotid atherosclerosis were performed using polarization filter reflected light microscopy to identify CC. In whole blood, CC exposure induced a time- and concentration-dependent generation of prothrombin fragment 1+2 (PTF1.2), tissue factor (TF) mRNA synthesis, and monocyte TF expression. Blocking Abs against TF abolished CC-mediated coagulation, thus indicating involvement of the TF-dependent pathway. Blockade of FXII by corn trypsin inhibitor had a significant inhibitory effect on CC-induced PTF1.2 in platelet-free plasma, although the overall activation potential was low. CC exposure did not induce platelet aggregation, TF microparticle induction, or TF on granulocytes or eosinophils. Inhibition of complement C3 by CP40 (compstatin), C5 by eculizumab, or C5aR1 by PMX53 blocked CC-induced PTF1.2 by 90% and reduced TF+ monocytes from 18-20 to 1-2%. The physiologic relevance was supported by birefringent CC structures adjacent to monocytes (CD14), TF, and activated complement iC3b and C5b-9 in a human brain thrombus. Furthermore, monocyte influx and TF induction in close proximity to CC-rich regions with activated complement were found in a vulnerable plaque. In conclusion, CC could be active, releasable contributors to thrombosis by inducing monocyte TF secondary to complement C5aR1 signaling.


Sujet(s)
Coagulation sanguine/immunologie , Cholestérol/immunologie , Activation du complément/immunologie , Récepteur à l'anaphylatoxine C5a/métabolisme , Thromboplastine/biosynthèse , Artériopathies carotidiennes/immunologie , Artériopathies carotidiennes/métabolisme , Humains , Monocytes/immunologie , Monocytes/métabolisme , Thromboplastine/immunologie , Thrombose/immunologie , Thrombose/métabolisme
19.
Front Immunol ; 10: 1209, 2019.
Article de Anglais | MEDLINE | ID: mdl-31214180

RÉSUMÉ

TLR8 is an endosomal sensor of RNA degradation products in human phagocytes, and is involved in the recognition of viral and bacterial pathogens. We previously showed that in human primary monocytes and monocyte derived macrophages, TLR8 senses entire Staphylococcus aureus and Streptococcus agalactiae (group B streptococcus, GBS), resulting in the activation of IRF5 and production of IFNß, IL-12p70, and TNF. However, the quantitative and qualitative impact of TLR8 for the sensing of bacteria have remained unclear because selective inhibitors have been unavailable. Moreover, while we have shown that TLR2 activation attenuates TLR8-IRF5 signaling, the molecular mechanism of this crosstalk is unknown. We here used a recently developed chemical antagonist of TLR8 to determine its role in human primary monocytes challenged with S. aureus, GBS, Streptococcus pneumonia, Pseudomonas aeruginosa, and E. coli. The inhibitor completely blocked cytokine production in monocytes stimulated with TLR8-agonists, but not TLR2-, and TLR4-agonists. Upon challenge with S. aureus, GBS, and S. pneumonia, the TLR8 inhibitor almost eliminated the production of IL-1ß and IL-12p70, and it strongly reduced the release of IL-6, TNF, and IL-10. With P. aeruginosa infection, the TLR8 inhibitor impaired the production of IL-12p70 and IL-1ß, while with E. coli infection the inhibitor had less effect that varied depending on the strain and conditions. Signaling via TLR2, TLR4, or TLR5, but not TLR8, rapidly eliminated IRAK-1 detection by immunoblotting due to IRAK-1 modifications during activation. Silencing of IRAK-1 reduced the induction of IFNß and TNF by TLR8 activation, suggesting that IRAK-1 is required for TLR8-IRF5 signaling. The TLR-induced modifications of IRAK-1 also correlated closely with attenuation of TLR8-IRF5 activation, suggesting that sequestration and/or modification of Myddosome components by cell surface TLRs limit the function of TLR8. Accordingly, inhibition of CD14- and TLR4-activation during E. coli challenge increased the activation of IRF5 and the production of IL-1ß and IL-12p70. We conclude that TLR8 is a dominating sensor of several species of pyogenic bacteria in human monocytes, while some bacteria attenuate TLR8-signaling via cell surface TLR- activation. Taken together, TLR8 appears as a more important sensor in the antibacterial defense system than previously known.


Sujet(s)
Macrophages/immunologie , Monocytes/immunologie , Infections à staphylocoques/immunologie , Staphylococcus aureus/physiologie , Streptococcus agalactiae/physiologie , Récepteur de type Toll-8/métabolisme , Cellules cultivées , Humains , Facteurs de régulation d'interféron/génétique , Facteurs de régulation d'interféron/métabolisme , Interféron bêta/métabolisme , Interleukin-1 Receptor-Associated Kinases/métabolisme , Interleukine-12/métabolisme , Transduction du signal , Facteur de nécrose tumorale alpha/métabolisme
20.
PLoS Pathog ; 15(3): e1007684, 2019 03.
Article de Anglais | MEDLINE | ID: mdl-30883606

RÉSUMÉ

Phagocytosis is a complex process that eliminates microbes and is performed by specialised cells such as macrophages. Toll-like receptor 4 (TLR4) is expressed on the surface of macrophages and recognizes Gram-negative bacteria. Moreover, TLR4 has been suggested to play a role in the phagocytosis of Gram-negative bacteria, but the mechanisms remain unclear. Here we have used primary human macrophages and engineered THP-1 monocytes to show that the TLR4 sorting adapter, TRAM, is instrumental for phagocytosis of Escherichia coli as well as Staphylococcus aureus. We find that TRAM forms a complex with Rab11 family interacting protein 2 (FIP2) that is recruited to the phagocytic cups of E. coli. This promotes activation of the actin-regulatory GTPases Rac1 and Cdc42. Our results show that FIP2 guided TRAM recruitment orchestrates actin remodelling and IRF3 activation, two events that are both required for phagocytosis of Gram-negative bacteria.


Sujet(s)
Protéines adaptatrices de la transduction du signal/physiologie , Protéines de transport/métabolisme , Protéines membranaires/métabolisme , Phagocytose/physiologie , Protéines adaptatrices de la transduction du signal/métabolisme , Animaux , Protéines de transport/physiologie , Endocytose , Endosomes , Escherichia coli/pathogénicité , Cellules HEK293 , Humains , Facteur-3 de régulation d'interféron , Lipopolysaccharides , Macrophages/immunologie , Macrophages/métabolisme , Protéines membranaires/physiologie , Souris , Souris de lignée C57BL , Facteur de différenciation myéloïde-88 , Culture de cellules primaires , Transport des protéines , Transduction du signal , Staphylococcus aureus/pathogénicité , Cellules THP-1 , Récepteur de type Toll-4/métabolisme , Protéine G cdc42 , Protéines G rab , Protéine G rac1
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE