Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Clin Immunol ; 247: 109240, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36693535

RESUMEN

Kruppel-like factor 2 (KLF2) has been linked with fibrosis and neutrophil-associated thromboinflammation; however, its role in COVID-19 remains elusive. We investigated the effect of disease microenvironment on the fibrotic potential of human lung fibroblasts (LFs) and its association with KLF2 expression. LFs stimulated with plasma from severe COVID-19 patients down-regulated KLF2 expression at mRNA/protein and functional level acquiring a pre-fibrotic phenotype, as indicated by increased CCN2/collagen levels. Pre-incubation with the COMBI-treatment-agents (DNase I and JAKs/IL-6 inhibitors baricitinib/tocilizumab) restored KLF2 levels of LFs to normal abolishing their fibrotic activity. LFs stimulated with plasma from COMBI-treated patients at day-7 expressed lower CCN2 and higher KLF2 levels, compared to plasma prior-to-treatment, an effect not observed in standard-of-care treatment. In line with this, COMBI-treated patients had better outcome than standard-of-care group. These data link fibroblast KLF2 with NETosis and JAK/IL-6 signaling, suggesting the potential of combined therapeutic strategies in immunofibrotic diseases, such as COVID-19.


Asunto(s)
COVID-19 , Factores de Transcripción de Tipo Kruppel , Trombosis , Humanos , Regulación hacia Abajo , Fibroblastos/metabolismo , Fibrosis , Inflamación , Interleucina-6/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Pulmón/metabolismo , Factores de Transcripción/genética
2.
Clin Exp Rheumatol ; 41(5): 1024-1033, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36135957

RESUMEN

OBJECTIVES: To assess the prevalence of autoantibodies (AAbs) in mechanically ventilated COVID-19 patients and to investigate whether AAbs influence the clinical outcome. METHODS: Serum samples were drawn within the first 48 hours upon admission to the intensive care unit (ICU) from 217 consecutive patients, from January 1st, 2021, to May 10th, 2021, and investigated for the presence of AAbs using conventional techniques. Serum samples (n=117) of age- and sex-matched healthy individuals collected before COVID-19 pandemic were used as controls. RESULTS: COVID-19 patients in the ICU had more commonly AAbs compared to age- and sex-matched controls (174/217, 80.2% vs. 73/117, 62.4%, p<0.001). Patients expressed more frequently ANAs (48.4% vs. 21.4%, p<0.001), anti-dsDNA (5.1% vs. 0%, p=0.01), anti-CCP (8.3% vs. 1.7%, p=0.014) and anti-CL IgM AAbs (21.7% vs. 9.4%, p=0.005) than controls, respectively. Simultaneous reactivity against at least three autoantigens, occurred in 144 out of 174 (82.8%) patients. The two groups did not differ in terms of clinicoepidemiologic characteristics or the mortality ratio within the ICU. Patients who died compared to convalescents were older, had higher ferritin, D-dimers levels, APACHE II score, lower oxygen saturation, higher prevalence of comorbidities and cognitive dysfunction. However, AAbs were not found to correlate with the clinical outcome. CONCLUSIONS: Patients with severe COVID-19 express AAbs more commonly compared to controls. No correlation was found between AAbs and disease outcome.


Asunto(s)
Autoanticuerpos , COVID-19 , Humanos , Autoinmunidad , Incidencia , Pandemias , COVID-19/epidemiología , Unidades de Cuidados Intensivos
3.
Clin Immunol ; 238: 109016, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35447311

RESUMEN

Aiming to reduce mortality in COVID-19 with severe respiratory failure we administered a combined rescue treatment (COMBI) on top of standard-of-care (SOC: dexamethasone/heparin) consisted of inhaled DNase to dissolve thrombogenic neutrophil extracellular traps, plus agents against cytokine-mediated hyperinflammation, namely anti-IL-6-receptor tocilizumab and JAK1/2 inhibitor baricitinib. Patients with PaO2/FiO2 < 100 mmHg were analysed. COMBI group (n = 22) was compared with similar groups that had received SOC alone (n = 26) or SOC plus monotherapy with either IL-1-receptor antagonist anakinra (n = 19) or tocilizumab (n = 11). COMBI was significantly associated with lower in-hospital mortality and intubation rate, shorter duration of hospitalization, and prolonged overall survival after a median follow-up of 110 days. In vitro, COVID-19 plasma induced tissue factor/thrombin pathway in primary lung fibroblasts. This effect was inhibited by the immunomodulatory agents of COMBI providing a mechanistic explanation for the clinical observations. These results support the conduct of randomized trials using combined immunomodulation in COVID-19 to target multiple interconnected pathways of immunothrombosis.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Tratamiento Farmacológico de COVID-19 , Desoxirribonucleasas , Insuficiencia Respiratoria , Anticuerpos Monoclonales Humanizados/uso terapéutico , Azetidinas/uso terapéutico , Desoxirribonucleasas/uso terapéutico , Humanos , Purinas/uso terapéutico , Pirazoles/uso terapéutico , Insuficiencia Respiratoria/tratamiento farmacológico , Insuficiencia Respiratoria/virología , SARS-CoV-2 , Sulfonamidas/uso terapéutico , Resultado del Tratamiento
4.
Eur J Immunol ; 51(4): 930-942, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33340091

RESUMEN

Ankylosing spondylitis (AS) is an inflammatory disease characterized by excessive bone formation. We investigated the presence of neutrophil extracellular traps (NETs) in AS and how they are involved in the osteogenic capacity of bone marrow mesenchymal stem cells (MSCs) through interleukin-17A (IL-17A). Peripheral neutrophils and sera were obtained from patients with active AS and healthy controls. NET formation and neutrophil/NET-associated proteins were studied using immunofluorescence, immunoblotting, qPCR, and ELISA. In vitro co-culture systems of AS NET structures and MSCs isolated from controls were deployed to examine the role of NETs in the differentiation of MSCs toward osteogenic cells. Analysis was performed using specific staining and qPCR. Neutrophils from patients with AS were characterized by enhanced formation of NETs carrying bioactive IL-17A and IL-1ß. IL-17A-enriched AS NETs mediated the differentiation of MSCs toward bone-forming cells. The neutrophil expression of IL-17A was positively regulated by IL-1ß. Blocking IL-1ß signaling on neutrophils with anakinra or dismantling NETs using DNase-I disrupted osteogenesis driven by IL-17A-bearing NETs. These findings propose a novel role of neutrophils in AS-related inflammation, linking IL-17A-decorated NETs with the differentiation of MSCs toward bone-forming cells. Moreover, IL-1ß triggers the expression of IL-17A on NETs offering an additional therapeutic target in AS.


Asunto(s)
Diferenciación Celular/inmunología , Trampas Extracelulares/inmunología , Interleucina-17/inmunología , Células Madre Mesenquimatosas/inmunología , Osteogénesis/inmunología , Adulto , Diferenciación Celular/genética , Células Cultivadas , Trampas Extracelulares/metabolismo , Femenino , Humanos , Interleucina-17/genética , Interleucina-17/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Persona de Mediana Edad , Neutrófilos/citología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Osteogénesis/genética , Espondilitis Anquilosante
5.
Rheumatology (Oxford) ; 61(4): 1639-1644, 2022 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-34260696

RESUMEN

OBJECTIVES: To explore the presence of neutrophil extracellular traps (NETs) in inflamed temporal artery biopsies (TABs) of patients with GCA. METHODS: Ten patients with GCA [five with limited and five with associated generalized vascular involvement, as defined by 18F-fluorodeoxyglucose PET with CT (PET/CT)] and eight with PMR were studied. The presence, location, quantitation and decoration of NETs with IL-6, IL-1ß and IL-17A were assessed in TABs at the time of disease diagnosis by tissue immunofluorescence and confocal microscopy. Paired serum levels of IL-6 and IL-17A were also evaluated in all patients. RESULTS: All temporal artery biopsies from GCA, but not PMR, patients had NETs located mainly in the adventitia, adjacent to the vasa vasorum. NETs decorated with IL-6 were present in 8/10 TABs of GCA patients, of whom 5 were PET/CT(+) and 3 PET/CT(-) patients. IL-17A(+) NETs were observed in all GCA patients. IL-1ß(+) NETs were not detected in any GCA patient. No relation was found between serum IL-6 and IL-17A levels and NETs containing IL-6 and/or IL-17A. CONCLUSIONS: NETs bearing pro-inflammatory cytokines are present in inflamed GCA-TABs. Future studies with a larger number of patients from different centres will show whether the findings regarding neutrophils/NETs in the TAB are consistent and disclose their clinical impact.


Asunto(s)
Trampas Extracelulares , Arteritis de Células Gigantes , Biopsia , Citocinas , Arteritis de Células Gigantes/diagnóstico , Humanos , Interleucina-17 , Interleucina-6 , Tomografía Computarizada por Tomografía de Emisión de Positrones , Arterias Temporales/diagnóstico por imagen , Arterias Temporales/patología
6.
Clin Immunol ; 227: 108750, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33945870

RESUMEN

The co-existence of an autoinflammatory syndrome with a demyelinating disorder is a very rare occurrence raising the question whether there is a pathophysiological connection between them. We describe the case of a man with symptoms of cryopyrin-associated periodic syndrome (CAPS) since infancy who later developed multiple sclerosis (MS). As CAPS was genetically confirmed, the inhibition of interleukin-1 (IL-1) with anakinra led to a swift resolution of the CAPS symptoms and also, in combination with teriflunomide, to a clinical and imaging improvement of MS. In vitro studies showed that, upon a CAPS flare, the patient's peripheral neutrophils released neutrophil extracellular traps (NETs) decorated with IL-1ß, while NET release was markedly decreased following anakinra-induced remission of CAPS. Taking into account the growing evidence on the involvement of IL-1ß in experimental models of MS, this rare patient case suggests that the role of neutrophils/NETs and IL-1ß in MS should be further studied.


Asunto(s)
Síndromes Periódicos Asociados a Criopirina/inmunología , Trampas Extracelulares/inmunología , Interleucina-1beta/inmunología , Esclerosis Múltiple/inmunología , Neutrófilos/inmunología , Adulto , Antirreumáticos/uso terapéutico , Encéfalo/diagnóstico por imagen , Crotonatos/uso terapéutico , Síndromes Periódicos Asociados a Criopirina/complicaciones , Síndromes Periódicos Asociados a Criopirina/tratamiento farmacológico , Síndromes Periódicos Asociados a Criopirina/fisiopatología , Humanos , Hidroxibutiratos/uso terapéutico , Inmunosupresores/uso terapéutico , Técnicas In Vitro , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Imagen por Resonancia Magnética , Masculino , Esclerosis Múltiple/complicaciones , Esclerosis Múltiple/diagnóstico por imagen , Esclerosis Múltiple/tratamiento farmacológico , Nitrilos/uso terapéutico , Toluidinas/uso terapéutico
7.
Int J Mol Sci ; 22(8)2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33921064

RESUMEN

Myelodysplastic syndromes (MDS) comprise a heterogeneous group of clonal hematopoietic stem (HSCs) and/or progenitor cells disorders. The established dependence of MDS progenitors on the hypoxic bone marrow (BM) microenvironment turned scientific interests to the transcription factor hypoxia-inducible factor 1 (HIF-1). HIF-1 facilitates quiescence maintenance and regulates differentiation by manipulating HSCs metabolism, being thus an appealing research target. Therefore, we examine the aberrant HIF-1 stabilization in BMs from MDS patients and controls (CTRLs). Using a nitroimidazole-indocyanine conjugate, we show that HIF-1 aberrant expression and transcription activity is oxygen independent, establishing the phenomenon of pseudohypoxia in MDS BM. Next, we examine mitochondrial quality and quantity along with levels of autophagy in the differentiating myeloid lineage isolated from fresh BM MDS and CTRL aspirates given that both phenomena are HIF-1 dependent. We show that the mitophagy of abnormal mitochondria and autophagic death are prominently featured in the MDS myeloid lineage, their severity increasing with intra-BM blast counts. Finally, we use in vitro cultured CD34+ HSCs isolated from fresh human BM aspirates to manipulate HIF-1 expression and examine its potential as a therapeutic target. We find that despite being cultured under 21% FiO2, HIF-1 remained aberrantly stable in all MDS cultures. Inhibition of the HIF-1α subunit had a variable beneficial effect in all <5%-intra-BM blasts-MDS, while it had no effect in CTRLs or in ≥5%-intra-BM blasts-MDS that uniformly died within 3 days of culture. We conclude that HIF-1 and pseudohypoxia are prominently featured in MDS pathobiology, and their manipulation has some potential in the therapeutics of benign MDS.


Asunto(s)
Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Hipoxia/fisiopatología , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/fisiopatología , Anciano , Anciano de 80 o más Años , Antígenos CD34/metabolismo , Autofagia/efectos de los fármacos , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mitofagia/efectos de los fármacos , Células Mieloides/efectos de los fármacos , Células Mieloides/metabolismo , Células Mieloides/ultraestructura , Nitroimidazoles/farmacología , Factores de Transcripción/metabolismo , Regulación hacia Arriba/efectos de los fármacos
8.
Clin Immunol ; 220: 108598, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32961333

RESUMEN

Growing clinical evidence has implicated complement as a pivotal driver of COVID-19 immunopathology. Deregulated complement activation may fuel cytokine-driven hyper-inflammation, thrombotic microangiopathy and NET-driven immunothrombosis, thereby leading to multi-organ failure. Complement therapeutics have gained traction as candidate drugs for countering the detrimental consequences of SARS-CoV-2 infection. Whether blockade of terminal complement effectors (C5, C5a, or C5aR1) may elicit similar outcomes to upstream intervention at the level of C3 remains debated. Here we compare the efficacy of the C5-targeting monoclonal antibody eculizumab with that of the compstatin-based C3-targeted drug candidate AMY-101 in small independent cohorts of severe COVID-19 patients. Our exploratory study indicates that therapeutic complement inhibition abrogates COVID-19 hyper-inflammation. Both C3 and C5 inhibitors elicit a robust anti-inflammatory response, reflected by a steep decline in C-reactive protein and IL-6 levels, marked lung function improvement, and resolution of SARS-CoV-2-associated acute respiratory distress syndrome (ARDS). C3 inhibition afforded broader therapeutic control in COVID-19 patients by attenuating both C3a and sC5b-9 generation and preventing FB consumption. This broader inhibitory profile was associated with a more robust decline of neutrophil counts, attenuated neutrophil extracellular trap (NET) release, faster serum LDH decline, and more prominent lymphocyte recovery. These early clinical results offer important insights into the differential mechanistic basis and underlying biology of C3 and C5 inhibition in COVID-19 and point to a broader pathogenic involvement of C3-mediated pathways in thromboinflammation. They also support the evaluation of these complement-targeting agents as COVID-19 therapeutics in large prospective trials.


Asunto(s)
Betacoronavirus/patogenicidad , Complemento C3/antagonistas & inhibidores , Complemento C5/antagonistas & inhibidores , Inactivadores del Complemento/uso terapéutico , Infecciones por Coronavirus/tratamiento farmacológico , Factores Inmunológicos/uso terapéutico , Neumonía Viral/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Biomarcadores/sangre , Proteína C-Reactiva/metabolismo , COVID-19 , Estudios de Cohortes , Activación de Complemento/efectos de los fármacos , Complemento C3/genética , Complemento C3/inmunología , Complemento C5/genética , Complemento C5/inmunología , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Trampas Extracelulares/efectos de los fármacos , Femenino , Expresión Génica , Humanos , Interleucina-6/metabolismo , Masculino , Persona de Mediana Edad , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/virología , Pandemias , Péptidos Cíclicos/uso terapéutico , Neumonía Viral/complicaciones , Neumonía Viral/inmunología , Neumonía Viral/virología , Síndrome de Dificultad Respiratoria/complicaciones , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/virología , SARS-CoV-2 , Índice de Severidad de la Enfermedad
9.
J Immunol ; 200(12): 3950-3961, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29712770

RESUMEN

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.


Asunto(s)
Autofagia/fisiología , Colitis Ulcerosa/metabolismo , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Neutrófilos/metabolismo , Factores de Transcripción/metabolismo , Adulto , Autofagia/efectos de los fármacos , Colitis Ulcerosa/tratamiento farmacológico , Colon/efectos de los fármacos , Colon/metabolismo , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/metabolismo , Trampas Extracelulares/efectos de los fármacos , Trampas Extracelulares/metabolismo , Femenino , Humanos , Inflamación/tratamiento farmacológico , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Masculino , Mesalamina/farmacología , Persona de Mediana Edad , Activación Neutrófila/efectos de los fármacos , Activación Neutrófila/fisiología , Neutrófilos/efectos de los fármacos
10.
Int J Mol Sci ; 21(10)2020 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-32455533

RESUMEN

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.


Asunto(s)
Trampas Extracelulares/efectos de los fármacos , Factores Inmunológicos/farmacología , Neutrófilos/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Ticagrelor/farmacología , Anciano , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Células Cultivadas , ADN/metabolismo , Trampas Extracelulares/metabolismo , Femenino , Humanos , Factores Inmunológicos/uso terapéutico , Masculino , Persona de Mediana Edad , Neutrófilos/inmunología , Peroxidasa/metabolismo , Inhibidores de Agregación Plaquetaria/uso terapéutico , Polifosfatos/metabolismo , Trombina/metabolismo , Ticagrelor/uso terapéutico
13.
Ann Rheum Dis ; 78(2): 238-248, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30563869

RESUMEN

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.


Asunto(s)
Trampas Extracelulares/metabolismo , Interleucina-17/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Tromboplastina/metabolismo , Factores de Transcripción/metabolismo , Autofagia/fisiología , Técnicas de Cultivo de Célula , Fibroblastos/metabolismo , Fibrosis/metabolismo , Humanos , Inflamación , Transducción de Señal , Trombosis/metabolismo
14.
J Autoimmun ; 104: 102305, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31337526

RESUMEN

Autoinflammatory disorders represent a heterogeneous group of systemic inflammatory diseases caused by genetic or acquired defects in key components of the innate immunity. Familial Mediterranean fever (FMF) is the most common among the other clinical phenotypes of the rare hereditary periodic fevers (HPFs) syndromes. FMF is associated with mutations in the MEFV gene encoding pyrin and is characterized by recurrent, often stress-provoked attacks of fever and serositis, but sometimes also by chronic subclinical inflammation. FMF is prevalent in Greece and other countries of the eastern Mediterranean region. Over the last 17 years, our group has focused on FMF as a model suitable for the research on innate immunity and particularly the role of neutrophils. Therefore, the study of Greek patients with FMF has yielded lessons across several levels: the epidemiology of the disease in Greece, the spectrum of its clinical manifestations and potential overlaps with other idiopathic inflammatory conditions, the demonstration of its rather complex and heterogeneous genetic background and the suggestion of a novel mechanism involved in the crosstalk between environmental stress and inflammation. Mechanistically, during FMF attack, neutrophils release chromatin structures called neutrophil extracellular traps (NETs), which are decorated with bioactive IL-1ß. REDD1 (regulated in development and DNA damage responses 1), that encodes a stress-related mTOR repressor, has been found to be the most significantly upregulated gene in neutrophils during disease attacks. Upon adrenergic stress, REDD1-induced autophagy triggers a pyrin-driven IL-1ß maturation, and the release of IL-1ß-bearing NETs. Consequently, not only the mode of action of IL-1ß-targeting therapies is explained, but also new treatment prospects emerge with the evaluation of old or the design of new drugs targeting autophagy-induced NETosis. Information gained from FMF studies may subsequently be applied in more complex but still relevant inflammatory conditions, such as adult-onset Still's disease, gout, ulcerative colitis and Behçet's disease.


Asunto(s)
Fiebre Mediterránea Familiar , Inmunidad Innata/genética , Modelos Inmunológicos , Mutación , Pirina , Fiebre Mediterránea Familiar/genética , Fiebre Mediterránea Familiar/inmunología , Fiebre Mediterránea Familiar/terapia , Humanos , Pirina/genética , Pirina/inmunología
15.
J Pathol ; 243(1): 111-122, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28678391

RESUMEN

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.


Asunto(s)
Coagulación Sanguínea , Plaquetas/metabolismo , Inflamación/sangre , Interleucinas/sangre , Neutrófilos/metabolismo , Polifosfatos/sangre , Infarto del Miocardio con Elevación del ST/sangre , Trombosis/sangre , Animales , Autofagia , Estudios de Casos y Controles , Cloruros , Modelos Animales de Enfermedad , Trampas Extracelulares/metabolismo , Compuestos Férricos , Humanos , Inflamación/inducido químicamente , Inflamación/prevención & control , Interferones , Interleucinas/administración & dosificación , Masculino , Ratones Endogámicos C57BL , Activación Plaquetaria , Infarto del Miocardio con Elevación del ST/diagnóstico por imagen , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Trombina/metabolismo , Trombosis/inducido químicamente , Trombosis/prevención & control
16.
J Allergy Clin Immunol ; 140(5): 1378-1387.e13, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28342915

RESUMEN

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.


Asunto(s)
Fiebre Mediterránea Familiar/inmunología , Inflamación/inmunología , Neutrófilos/inmunología , Estrés Psicológico/inmunología , Factores de Transcripción/metabolismo , Adulto , Autofagia , Progresión de la Enfermedad , Trampas Extracelulares/metabolismo , Fiebre Mediterránea Familiar/genética , Femenino , Humanos , Interleucina-1beta/metabolismo , Masculino , Pirina/genética , Remisión Espontánea , Estrés Fisiológico/inmunología , Adulto Joven
17.
Eur J Immunol ; 46(11): 2542-2554, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27585946

RESUMEN

Aberrant formation of neutrophil extracellular traps (NETs) is a key feature in rheumatoid arthritis (RA) and plays a pivotal role in disease pathogenesis. However, the mechanism through which NETs shape the autoimmune response in RA remains elusive. In this study, we demonstrate that inhibition of peptidylarginine deiminases activity in collagen-induced arthritis (CIA) mouse model significantly reduces NET formation, attenuates clinical disease activity, and prevents joint destruction. Importantly, peptidylarginine deiminase 4 blocking markedly reduces the frequency of collagen-specific IFN-γ-producing T helper 1 (Th1) cells in the draining lymph nodes of immunized mice. Exposure of dendritic cells (DCs) to CIA-derived NETs induces DC maturation characterized by significant upregulation of costimulatory molecules, as well as elevated secretion of IL-6. Moreover, CIA-NET-treated DCs promote the induction of antigen-specific Th1 cells in vitro. Finally, NETs from RA patients show an increased potential to induce the maturation of DCs from healthy individuals, corroborating the findings obtained in CIA mouse model. Collectively, our findings delineate an important role of NETs in the induction and expansion of Th1 pathogenic cells in CIA through maturation of DCs and reveal a novel role of NETs in shaping the RA-autoimmune response that could be exploited therapeutically.


Asunto(s)
Artritis Reumatoide/inmunología , Autoinmunidad , Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Trampas Extracelulares/inmunología , Células TH1/inmunología , Animales , Artritis Experimental/inmunología , Artritis Reumatoide/fisiopatología , Colágeno/administración & dosificación , Colágeno/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Trampas Extracelulares/efectos de los fármacos , Humanos , Hidrolasas/metabolismo , Interferón gamma/metabolismo , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos DBA , Ornitina/administración & dosificación , Ornitina/análogos & derivados , Arginina Deiminasa Proteína-Tipo 4
18.
Antimicrob Agents Chemother ; 60(2): 1040-8, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26643338

RESUMEN

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.


Asunto(s)
Infecciones por Acinetobacter/tratamiento farmacológico , Claritromicina/farmacología , Trampas Extracelulares/efectos de los fármacos , Factores Inmunológicos/farmacología , Neutrófilos/efectos de los fármacos , Infecciones por Acinetobacter/inmunología , Infecciones por Acinetobacter/patología , Acinetobacter baumannii/patogenicidad , Adulto , Antibacterianos/inmunología , Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos , Autofagia/efectos de los fármacos , Estudios de Casos y Controles , Catelicidinas/metabolismo , Claritromicina/inmunología , Femenino , Gastritis/sangre , Infecciones por Helicobacter/sangre , Infecciones por Helicobacter/microbiología , Humanos , Factores Inmunológicos/inmunología , Masculino , Persona de Mediana Edad , Neutrófilos/inmunología , Neutrófilos/patología
19.
Ann Rheum Dis ; 75(1): 269-77, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25261578

RESUMEN

OBJECTIVE: Inflammatory attacks of familial Mediterranean fever (FMF) are characterised by circulation and influx of high number of polymorphonuclear neutrophils (PMN) in the affected sites and profound therapeutic effect of IL-1ß inhibitors. We investigated the role of neutrophil extracellular traps (NET) in the pathogenesis of FMF, and their involvement in IL-1ß production. METHODS: Blood samples were obtained from six FMF patients during remissions and from three patients during attacks. NET formation and NET components were studied by fluorescence techniques, immunobloting and MPO-DNA complex ELISA. RESULTS: PMNs from patients released NETs decorated with IL-1ß during disease attacks. On the other hand, PMNs from patients during remission were resistant to inflammatory stimuli that induce NET release in PMNs from control subjects. Lower basal autophagy levels were identified in PMNs during remission, while induction of autophagy facilitated NET release, suggesting that autophagy is involved in the regulation of NET release. During the resolution of attacks, inhibition of NET formation by negative feedback mechanism was also observed. The anti-inflammatory agents, colchicine and DNAse I, inhibited IL-1ß production in PMNs and IL-1ß activity in NETs, respectively. CONCLUSIONS: We suggest two additive events for triggering the FMF attack; the production of IL-1ß by PMNs and its release through NETs. At the same time NETs, homeostatically, downregulate further NETosis, facilitating the resolution of attack. Compensatorly, lower basal autophagy of PMNs may protect from crises by attenuating the release of pro-inflammatory NETs.


Asunto(s)
Trampas Extracelulares/inmunología , Fiebre Mediterránea Familiar/inmunología , Interleucina-1beta/inmunología , Neutrófilos/inmunología , Adulto , Antiinflamatorios/farmacología , Autofagia/inmunología , Estudios de Casos y Controles , Colchicina/farmacología , Desoxirribonucleasa I/farmacología , Trampas Extracelulares/efectos de los fármacos , Retroalimentación Fisiológica , Femenino , Humanos , Interleucina-1beta/biosíntesis , Masculino , Persona de Mediana Edad , Neutrófilos/efectos de los fármacos , Inducción de Remisión , Adulto Joven
20.
Eur Heart J ; 36(22): 1405-14, 2015 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-25660055

RESUMEN

AIMS: Neutrophil extracellular traps (NETs) are chromatin filaments released by activated polymorphonuclear neutrophils (PMNs) and decorated with granule proteins with various properties. Several lines of evidence implicate NETs in thrombosis. The functional significance and the in vivo relevance of NETs during atherothrombosis in humans have not been addressed until now. METHODS AND RESULTS: Selective sampling of thrombotic material and surrounding blood from the infarct-related coronary artery (IRA) and the non-IRA was performed during primary percutaneous revascularization in 18 patients with ST-segment elevation acute myocardial infarction (STEMI). Thrombi isolated from IRA contained PMNs and NETs decorated with tissue factor (TF). Although TF was expressed intracellularly in circulating PMNs of STEMI patients, active TF was specifically exposed by NETs obtained from the site of plaque rupture. Treatment of NET structures with DNase I abolished TF functionality measurement. In vitro treatment of control PMNs with plasma obtained from IRA and non-IRA was further shown to induce intracellular up-regulation of TF but not NET formation. A second step consisting of the interaction between PMNs and thrombin-activated platelets was required for NET generation and subsequent TF exposure. CONCLUSION: The interaction of thrombin-activated platelets with PMNs at the site of plaque rupture during acute STEMI results in local NET formation and delivery of active TF. The notion that NETs represent a mechanism by which PMNs release thrombogenic signals during atherothrombosis may offer novel therapeutic targets.


Asunto(s)
Vasos Coronarios/metabolismo , Trampas Extracelulares/metabolismo , Infarto del Miocardio/metabolismo , Neutrófilos/metabolismo , Tromboplastina/metabolismo , Análisis de Varianza , Estudios de Casos y Controles , Trombosis Coronaria/metabolismo , Trombosis Coronaria/cirugía , Femenino , Humanos , Leucocitos Mononucleares/fisiología , Masculino , Persona de Mediana Edad , Infarto del Miocardio/cirugía , Revascularización Miocárdica/métodos , Intervención Coronaria Percutánea , Placa Aterosclerótica , Activación Plaquetaria/fisiología , Rotura Espontánea/metabolismo , Trombina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA