Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 21(10)2020 May 21.
Article in English | MEDLINE | ID: mdl-32455533

ABSTRACT

Neutrophils through the release of neutrophil extracellular traps (NETs) containing active tissue factor (TF) are key components of thrombo-inflammation. Platelets-neutrophils interplay in ST elevation myocardial infarction (STEMI) promotes NET formation via inorganic polyphosphates (polyP) released by thrombin-activated platelets. NETs, however, are also induced by biomaterials in a platelet-independent manner. Considering the possible pleiotropic effects of Ticagrelor beyond platelet inhibition and the clinical need for novel antithrombotic strategies targeting inflammation, we investigated the effects of Ticagrelor on polyP and stent-induced NETs in STEMI. Neutrophils from healthy individuals and patients receiving Ticagrelor were stimulated with polyP or drug-eluting stents (DES) to produce NETs. To induce TF expression, neutrophils were further incubated with plasma obtained from the infarct-related artery (IRA) of STEMI patients. The effects of Ticagrelor on NETs and TF loading were assessed using fluorescence microscopy, flow cytometry, myeloperoxidase(MPO)/DNA complex ELISA, and a Western blot. Ticagrelor interrupts platelet-neutrophil interaction by attenuating NETs induced by polyP. However, Ticagrelor does not affect polyP secretion from thrombin-activated platelets. Similarly, the intracellular production of TF in neutrophils triggered by IRA plasma is not hindered by Ticagrelor. Furthermore, DES induce NETs and synchronous stimulation with IRA plasma leads to the formation of thrombogenic TF-bearing NETs. Ticagrelor inhibits stent-induced NET release. These findings suggest a novel immune-modulatory effect of Ticagrelor when it attenuates the formation of thrombogenic NETs.


Subject(s)
Extracellular Traps/drug effects , Immunologic Factors/pharmacology , Neutrophils/drug effects , Platelet Aggregation Inhibitors/pharmacology , Ticagrelor/pharmacology , Aged , Blood Platelets/drug effects , Blood Platelets/metabolism , Cells, Cultured , DNA/metabolism , Extracellular Traps/metabolism , Female , Humans , Immunologic Factors/therapeutic use , Male , Middle Aged , Neutrophils/immunology , Peroxidase/metabolism , Platelet Aggregation Inhibitors/therapeutic use , Polyphosphates/metabolism , Thrombin/metabolism , Ticagrelor/therapeutic use
2.
Ann Rheum Dis ; 78(2): 238-248, 2019 02.
Article in English | MEDLINE | ID: mdl-30563869

ABSTRACT

OBJECTIVES: The release of neutrophil extracellular traps (NETs) represents a novel neutrophil effector function in systemic lupus erythematosus (SLE) pathogenesis. However, the molecular mechanism underlying NET release and how NETs mediate end-organ injury in SLE remain elusive. METHODS: NET formation and NET-related proteins were assessed in the peripheral blood and biopsies from discoid lupus and proliferative nephritis, using immunofluorescence, immunoblotting, quantitative PCR and ELISA. Autophagy was assessed by immunofluorescence and immunoblotting. The functional effects of NETs in vitro were assessed in a primary fibroblast culture. RESULTS: Neutrophils from patients with active SLE exhibited increased basal autophagy levels leading to enhanced NET release, which was inhibited in vitro by hydroxychloroquine. NETosis in SLE neutrophils correlated with increased expression of the stress-response protein REDD1. Endothelin-1 (ET-1) and hypoxia-inducible factor-1α (HIF-1α) were key mediators of REDD1-driven NETs as demonstrated by their inhibition with bosentan and L-ascorbic acid, respectively. SLE NETs were decorated with tissue factor (TF) and interleukin-17A (IL-17A), which promoted thrombin generation and the fibrotic potential of cultured skin fibroblasts. Notably, TF-bearing and IL-17A-bearing NETs were abundant in discoid skin lesions and in the glomerular and tubulointerstitial compartment of proliferative nephritis biopsy specimens. CONCLUSIONS: Our data suggest the involvement of REDD1/autophagy/NET axis in end-organ injury and fibrosis in SLE, a likely candidate for repositioning of existing drugs for SLE therapy. Autophagy-mediated release of TF-bearing and IL-17A-bearing NETs provides a link between thromboinflammation and fibrosis in SLE and may account for the salutary effects of hydroxychloroquine.


Subject(s)
Extracellular Traps/metabolism , Interleukin-17/metabolism , Lupus Erythematosus, Systemic/metabolism , Thromboplastin/metabolism , Transcription Factors/metabolism , Autophagy/physiology , Cell Culture Techniques , Fibroblasts/metabolism , Fibrosis/metabolism , Humans , Inflammation , Signal Transduction , Thrombosis/metabolism
3.
Front Immunol ; 9: 2064, 2018.
Article in English | MEDLINE | ID: mdl-30250474

ABSTRACT

Background: Type 2 diabetes mellitus (T2D) is characterized by susceptibility to bacterial infections and impaired wound healing. Neutrophil extracellular traps (NETs) and the cathelicidin antimicrobial peptide LL-37 have been implicated both in defense against bacterial infections and in wound healing process. Recently, it was shown that macrolide antibiotic clarithromycin induces the release of LL-37-bearing NETs. In T2D there has not been identified any link between NETs and LL-37 and the effect of clarithromycin in neutrophils/NETs is unknown yet. Methods: Peripheral blood neutrophils were obtained from treatment-naive hyperglycemic T2D patients (naive), normoglycemic T2D patients under antidiabetic treatment (well-controlled) and healthy donors (controls). NET release and NET proteins were studied. Co-culture systems of NET structures with E. coli NCTC 9001 and primary skin fibroblasts were deployed to examine the in vitro antibacterial and fibrotic NET properties, respectively. The effect of clarithromycin was also investigated. Analysis was performed using immunofluorescence confocal microscopy, myeloperoxidase-DNA complex and LL-37 ELISA, immunoblotting and qRT-PCR. Results: NETs were characterized by the presence of LL-37, however they lacked antibacterial activity, in both groups of T2D patients. Clarithromycin significantly increased the externalization of LL-37 on NETs generated from well-controlled T2D neutrophils, thus restoring NET antibacterial capacity and promoting the wound healing process via fibroblast activation and differentiation. Conclusion: This study suggests that clarithromycin may add further advantage to well-controlled T2D patients, by enhancing their antibacterial defense and improving wound healing capacity of fibroblasts, through upregulation of LL-37 on NET structures.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Antimicrobial Cationic Peptides/metabolism , Clarithromycin/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Extracellular Traps/metabolism , Fibroblasts/pathology , Neutrophils/immunology , Aged , Cell Differentiation , Cells, Cultured , Coculture Techniques , Female , Fibrosis , Humans , Male , Middle Aged , Wound Healing/drug effects , Cathelicidins
4.
J Immunol ; 200(12): 3950-3961, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29712770

ABSTRACT

Infiltration of neutrophils into colonic mucosa has been associated with the severity of ulcerative colitis (UC). We investigated the effect of disease microenvironment on the release of neutrophil extracellular traps (NETs) as well as the involved mechanisms in NETosis and whether certain NET proteins are correlated with disease phenotype. Peripheral blood neutrophils, sera, and colonic tissue were collected from treatment-naive and mesalazine-treated patients with active UC, treatment-naive patients with active Crohn's disease, patients suffering from infectious colitis, or healthy individuals (controls). Analysis of colonic biopsy specimens and peripheral blood neutrophils for the presence of NET-related markers using immunofluorescence confocal microscopy, ELISA, immunoblotting, flow cytometry, and quantitative PCR were performed. In vitro cell and tissue culture systems were further deployed. The local inflammatory response in colon in UC, but not Crohn's disease, is characterized by the presence of NETs carrying bioactive IL-1ß and thrombogenic tissue factor. The inflammatory environment of UC is able to induce neutrophil activation, IL-1ß expression, and NET release, as shown both ex vivo and in vitro. REDD1 expression, as a mediator linking inflammation, autophagy, and NET release, was also specifically associated with the inflammatory response of UC. We show that neutrophil expression of REDD1 in colon tissue and the presence of IL-1ß in neutrophils/NETs provide candidate biomarkers for the differential diagnosis of inflammatory colitis and possible targets for the treatment of UC, suggesting that UC shares common features with autoinflammatory disorders.


Subject(s)
Autophagy/physiology , Colitis, Ulcerative/metabolism , Inflammation/metabolism , Interleukin-1beta/metabolism , Neutrophils/metabolism , Transcription Factors/metabolism , Adult , Autophagy/drug effects , Colitis, Ulcerative/drug therapy , Colon/drug effects , Colon/metabolism , Crohn Disease/drug therapy , Crohn Disease/metabolism , Extracellular Traps/drug effects , Extracellular Traps/metabolism , Female , Humans , Inflammation/drug therapy , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Mesalamine/pharmacology , Middle Aged , Neutrophil Activation/drug effects , Neutrophil Activation/physiology , Neutrophils/drug effects
5.
J Pathol ; 243(1): 111-122, 2017 09.
Article in English | MEDLINE | ID: mdl-28678391

ABSTRACT

Neutrophils and neutrophil-released meshwork structures termed neutrophil extracellular traps (NETs) are major mediators of thromboinflammation and emerging targets for therapy, yet the mechanisms and pathways that control the role of neutrophils in thromboinflammation remain poorly understood. Here, we explored the role of IFN-λ1/IL-29, a major antiviral cytokine recently shown to suppress the neutrophil migratory capacity, in prothrombotic and proNETotic functions of neutrophils. In an ex vivo human experimental setting of acute ST-segment elevation myocardial infarction (STEMI), we show that IFN-λ1/IL-29 hinders NET release and diminishes the amount of cytoplasmic TF in neutrophils. Since platelet-neutrophil interaction plays a major role in NET-induced thromboinflammation, we further studied how IFN-λ1/IL-29 may interrupt this interaction. In this context, we identified inorganic polyphosphate (polyP) as a platelet-derived NET inducer in STEMI. In arterial STEMI thrombi, polyP was present in platelets and in close proximity to NET remnants. PolyP release from activated platelets was dependent on thrombin present in infarcted artery plasma, resulting in NET formation by promoting mTOR inhibition and autophagy induction. The effect of polyP on mTOR inhibition was counteracted by IFN-λ1/IL-29 treatment, leading to inhibition of NET formation. Consistently, we show in an in vivo model of FeCl3 -induced arterial thrombosis that IFN-λ2/IL-28A exerts strong antithrombotic potential. Taken together, these findings reveal a novel function of IFN-λ1/IL-29 in the suppression of thromboinflammation. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Blood Coagulation , Blood Platelets/metabolism , Inflammation/blood , Interleukins/blood , Neutrophils/metabolism , Polyphosphates/blood , ST Elevation Myocardial Infarction/blood , Thrombosis/blood , Animals , Autophagy , Case-Control Studies , Chlorides , Disease Models, Animal , Extracellular Traps/metabolism , Ferric Compounds , Humans , Inflammation/chemically induced , Inflammation/prevention & control , Interferons , Interleukins/administration & dosage , Male , Mice, Inbred C57BL , Platelet Activation , ST Elevation Myocardial Infarction/diagnostic imaging , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Thrombin/metabolism , Thrombosis/chemically induced , Thrombosis/prevention & control
6.
J Allergy Clin Immunol ; 140(5): 1378-1387.e13, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28342915

ABSTRACT

BACKGROUND: Familial Mediterranean fever (FMF) is an IL-1ß-dependent autoinflammatory disease caused by mutations of Mediterranean fever (MEFV) encoding pyrin and characterized by inflammatory attacks induced by physical or psychological stress. OBJECTIVE: We investigated the underlying mechanism that links stress-induced inflammatory attacks with neutrophil activation and release of IL-1ß-bearing neutrophil extracellular traps (NETs) in patients with FMF. METHODS: RNA sequencing was performed in peripheral neutrophils from 3 patients with FMF isolated both during attacks and remission, 8 patients in remission, and 8 healthy subjects. NET formation and proteins were analyzed by using confocal immunofluorescence microscopy, immunoblotting, myeloperoxidase-DNA complex ELISA, and flow cytometry. Samples from patients with Still's disease and bacterial infections were used also. RESULTS: The stress-related protein regulated in development and DNA damage responses 1 (REDD1) is significantly overexpressed during FMF attacks. Neutrophils from patients with FMF during remission are resistant to autophagy-mediated NET release, which can be overcome through REDD1 induction. Stress-related mediators (eg, epinephrine) decrease this threshold, leading to autophagy-driven NET release, whereas the synchronous inflammatory environment of FMF attack leads to intracellular production of IL-1ß and its release through NETs. REDD1 in autolysosomes colocalizes with pyrin and nucleotide-binding domain, leucine-rich repeat/pyrin domain-containing 3. Mutated pyrin prohibits this colocalization, leading to higher IL-1ß levels on NETs. CONCLUSIONS: This study provides a link between stress and initiation of inflammatory attacks in patients with FMF. REDD1 emerges as a regulator of neutrophil function upstream to pyrin, is involved in NET release and regulation of IL-1ß, and might constitute an important piece in the IL-1ß-mediated inflammation puzzle.


Subject(s)
Familial Mediterranean Fever/immunology , Inflammation/immunology , Neutrophils/immunology , Stress, Psychological/immunology , Transcription Factors/metabolism , Adult , Autophagy , Disease Progression , Extracellular Traps/metabolism , Familial Mediterranean Fever/genetics , Female , Humans , Interleukin-1beta/metabolism , Male , Pyrin/genetics , Remission, Spontaneous , Stress, Physiological/immunology , Young Adult
7.
PLoS One ; 11(5): e0154484, 2016.
Article in English | MEDLINE | ID: mdl-27136460

ABSTRACT

BACKGROUND: The role of neutrophils in tumour biology is largely unresolved. Recently, independent studies indicated either neutrophil extracellular traps (NETs) or Tissue Factor (TF) involvement in cancer biology and associated thrombosis. However, their individual or combined role in colonic adenocarcinoma is still unexplored. METHODS: Colectomy tissue specimens and variable number of draining lymph nodes were obtained from ten patients with adenocarcinoma of the colon. NETs deposition and neutrophil presence as well as TF expression were examined by immunostaining. The effect of NETs on cancer cell growth was studied in in vitro co-cultures of Caco-2 cell line and acute myeloid leukemia primary cells. Proliferation and apoptosis/necrosis of cancer cells were analyzed by flow cytometry. RESULTS: TF-bearing NETs and neutrophil localization were prominent in tumour sections and the respective metastatic lymph nodes. Interestingly, neutrophil infiltration and NETs concentration were gradually reduced from the tumour mass to the distal margin. The in vitro-generated NETs impeded growth of cancer cell cultures by inducing apoptosis and/or inhibiting proliferation. CONCLUSIONS: These data support further the role of neutrophils and NETs in cancer biology. We also suggest their involvement on cancer cell growth.


Subject(s)
Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Neutrophil Infiltration/immunology , Neutrophil Infiltration/physiology , Neutrophils/immunology , Neutrophils/physiology , Caco-2 Cells , Extracellular Traps/immunology , Extracellular Traps/physiology , Flow Cytometry , Humans , Tumor Cells, Cultured
8.
Antimicrob Agents Chemother ; 60(2): 1040-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26643338

ABSTRACT

Macrolide antibiotics have been shown to act as immunomodulatory molecules in various immune cells. However, their effect on neutrophils has not been extensively investigated. In this study, we investigated the role of macrolide antibiotics in the generation of neutrophil extracellular traps (NETs). By assessing ex vivo and in vivo NET formation, we demonstrated that clarithromycin is able to induce NET generation both in vitro and in vivo. Clarithromycin utilizes autophagy in order to form NETs, and these NETs are decorated with antimicrobial peptide LL-37. Clarithromycin-induced NETs are able to inhibit Acinetobacter baumannii growth and biofilm formation in an LL-37-dependent manner. Additionally, LL-37 antimicrobial function depends on NET scaffold integrity. Collectively, these data expand the knowledge on the immunomodulatory role of macrolide antibiotics via the generation of LL-37-bearing NETs, which demonstrate LL-37-dependent antimicrobial activity and biofilm inhibition against A. baumannii.


Subject(s)
Acinetobacter Infections/drug therapy , Clarithromycin/pharmacology , Extracellular Traps/drug effects , Immunologic Factors/pharmacology , Neutrophils/drug effects , Acinetobacter Infections/immunology , Acinetobacter Infections/pathology , Acinetobacter baumannii/pathogenicity , Adult , Anti-Bacterial Agents/immunology , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides , Autophagy/drug effects , Case-Control Studies , Cathelicidins/metabolism , Clarithromycin/immunology , Female , Gastritis/blood , Helicobacter Infections/blood , Helicobacter Infections/microbiology , Humans , Immunologic Factors/immunology , Male , Middle Aged , Neutrophils/immunology , Neutrophils/pathology
9.
J Pediatr Endocrinol Metab ; 27(3-4): 221-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24150199

ABSTRACT

BACKGROUND: Plasma adipocytokines are associated with metabolic profile and cardiovascular risk in obese children. OBJECTIVE: To investigate the association of plasma leptin and adiponectin concentrations with cardiometabolic risk profile and systemic inflammation in non-obese children. SUBJECTS: We studied 170 healthy, non-obese children (86 males, mean age 10±2 years). METHODS: Children's current body mass index (BMI), plasma leptin and adiponectin concentrations, lipid profile, fasting plasma glucose and high sensitivity C reactive protein (hsCRP) were measured. RESULTS: After adjustment for age, gender and BMI, plasma leptin concentrations were positively associated with hsCRP (t=2.72, p=0.009) and fasting plasma glucose (t=4.27, p<0.0001); plasma adiponectin concentrations were negatively associated with hsCRP (t=-3.31, p=0.0016); and positively with high density lipoprotein cholesterol (t=2.32, p=0.02). Children in the highest quartile of leptin/adiponectin (L/A) ratio demonstrated significantly higher BMI, systolic blood pressure, hsCRP, triglycerides and fasting glucose and the lowest high density lipoprotein (HDL) compared to lower L/A ratio quartiles. CONCLUSIONS: Alterations in plasma leptin and adiponectin may help to reclassify non-obese children, detecting those with more unfavorable risk profiles independent of BMI status.


Subject(s)
Adiponectin/blood , Inflammation/blood , Leptin/blood , Metabolic Diseases/blood , Body Mass Index , Child , Female , Healthy Volunteers , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...