Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
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 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
3.
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
4.
Int J Mol Sci ; 23(24)2022 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-36555464

RESUMEN

Several studies have indicated that a neutrophil extracellular trap (NET) formation, apart from its role in host defense, can contribute to or drive pathogenesis in a wide range of inflammatory and thrombotic disorders. Therefore, NETs may serve as a therapeutic target or/and a diagnostic tool. Here, we compare the most commonly used techniques for the assessment of NET formation. Furthermore, we review recent data from the literature on the application of basic laboratory tools for detecting NET release and discuss the challenges and the advantages of these strategies in NET evaluation. Taken together, we provide some important insights into the qualitative and quantitative molecular analysis of NETs in translational medicine today.


Asunto(s)
Trampas Extracelulares , Neutrófilos , Investigación Biomédica Traslacional , Ciencia Traslacional Biomédica , Biología
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
J Pathol ; 233(3): 294-307, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24740698

RESUMEN

Neutrophil activation by inflammatory stimuli and the release of extracellular chromatin structures (neutrophil extracellular traps - NETs) have been implicated in inflammatory disorders. Herein, we demonstrate that NETs released by neutrophils treated either with fibrosis-related agents, such as cigarette smoke, magnesium silicate, bleomycin, or with generic NET inducers, such as phorbol 12-myristate 13-acetate, induced activation of lung fibroblasts (LFs) and differentiation into myofibroblast (MF) phenotype. Interestingly, the aforementioned agents or IL-17 (a primary initiator of inflammation/fibrosis) had no direct effect on LF activation and differentiation. MFs treated with NETs demonstrated increased connective tissue growth factor expression, collagen production, and proliferation/migration. These fibrotic effects were significantly decreased after degradation of NETs with DNase1, heparin or myeloperoxidase inhibitor, indicating the key role of NET-derived components in LF differentiation and function. Furthermore, IL-17 was expressed in NETs and promoted the fibrotic activity of differentiated LFs but not their differentiation, suggesting that priming by DNA and histones is essential for IL-17-driven fibrosis. Additionally, autophagy was identified as the orchestrator of NET formation, as shown by inhibition studies using bafilomycin A1 or wortmannin. The above findings were further supported by the detection of NETs in close proximity to alpha-smooth muscle actin (α-SMA)-expressing fibroblasts in biopsies from patients with fibrotic interstitial lung disease or from skin scar tissue. Together, these data suggest that both autophagy and NETs are involved not only in inflammation but also in the ensuing fibrosis and thus may represent potential therapeutic targets in human fibrotic diseases.


Asunto(s)
Diferenciación Celular , Cromatina/metabolismo , Cicatriz/metabolismo , Pulmón/metabolismo , Miofibroblastos/metabolismo , Neutrófilos/metabolismo , Comunicación Paracrina , Fibrosis Pulmonar/metabolismo , Actinas/metabolismo , Autofagia , Diferenciación Celular/efectos de los fármacos , Movimiento Celular , Proliferación Celular , Células Cultivadas , Cicatriz/inmunología , Cicatriz/patología , Técnicas de Cocultivo , Colágeno/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Inhibidores Enzimáticos/farmacología , Fibrosis , Humanos , Interleucina-17/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Miofibroblastos/efectos de los fármacos , Miofibroblastos/patología , Activación Neutrófila , Infiltración Neutrófila , Neutrófilos/efectos de los fármacos , Comunicación Paracrina/efectos de los fármacos , Peroxidasa/metabolismo , Fenotipo , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/patología , Piel/inmunología , Piel/metabolismo , Piel/patología
14.
Ann Rheum Dis ; 73(10): 1854-63, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23873874

RESUMEN

OBJECTIVES: Antineutrophil cytoplasmic antibody (ANCA) associated vasculitis (AAV) is characterised by neutrophil activation. An elevated prevalence of venous thromboembolic events has been reported in AAV. Because of the critical role of neutrophils in inflammation associated thrombosis, we asked whether neutrophil tissue factor (TF) may be implicated in the thrombotic diathesis in AAV. METHODS: Neutrophils from four patients and sera from 17 patients with ANCA associated vasculitis with active disease and remission were studied. TF expression was assessed by immunoblotting and confocal microscopy. Circulating DNA levels were evaluated. TF expressing microparticles (MPs) were measured by flow cytometry and thrombin-antithrombin complex levels by ELISA. RESULTS: Peripheral blood neutrophils from four patients with active disease expressed elevated TF levels and released TF expressing neutrophil extracellular traps (NETs) and MPs. TF positive NETs were released by neutrophils isolated from the bronchoalveolar lavage and were detected in nasal and renal biopsy specimens. Elevated levels of circulating DNA and TF expressing neutrophil derived MPs were further observed in sera from patients with active disease. Induction of remission attenuated the aforementioned effects. Control neutrophils treated with sera from patients with active disease released TF bearing NETs and MPs which were abolished after IgG depletion. Treatment of control neutrophils with isolated IgG from sera from patients with active disease also resulted in the release of TF bearing NETs. TF implication in MP dependent thrombin generation was demonstrated by antibody neutralisation studies. CONCLUSIONS: Expression of TF in NETs and neutrophil derived MPs proposes a novel mechanism for the induction of thrombosis and inflammation in active AAV.


Asunto(s)
Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/sangre , Micropartículas Derivadas de Células/metabolismo , Trampas Extracelulares/metabolismo , Neutrófilos/metabolismo , Trombofilia/etiología , Tromboplastina/fisiología , Adulto , Anciano , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/complicaciones , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/terapia , Anticuerpos Anticitoplasma de Neutrófilos/sangre , Líquido del Lavado Bronquioalveolar/citología , Estudios de Casos y Controles , Femenino , Humanos , Inmunoglobulina G/inmunología , Masculino , Persona de Mediana Edad , Activación Neutrófila , Inducción de Remisión , Trombofilia/metabolismo , Tromboplastina/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
15.
J Immunol ; 186(11): 6568-75, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21531894

RESUMEN

Neonatal respiratory distress syndrome can progress to bronchopulmonary dysplasia (BPD), a serious pulmonary fibrotic disorder. Given the involvement of the extrinsic coagulation cascade in animal models of lung fibrosis, we examined its role in BPD. We observed a higher number of neutrophils expressing tissue factor (TF) in bronchoalveolar lavage fluid (BALF) from infants with BPD than from those with uncomplicated respiratory distress syndrome together with a parallel decrease in TF and connective tissue growth factor (CTGF) in BALF supernatants during the disease course. The involvement of coagulation in the fibrotic process associated with BPD was further evaluated by treating primary human colonic myofibroblasts with BALF supernatants from infants with BPD. These human colonic myofibroblasts demonstrated an enhanced C5a- and thrombin-dependent migration. Moreover, they expressed TF in an endothelin-1-dependent manner, with subsequent activation of the extrinsic coagulation cascade and CTGF production mediated by protease-activator receptor-1 signaling. These data provide a novel mechanism for the development of BPD and indicate that endothelin-1 signaling contributes to fibrosis by upregulating a TF/thrombin amplification loop responsible for CTGF production, and offer novel and specific therapeutic targets for pulmonary fibrotic disease.


Asunto(s)
Displasia Broncopulmonar/metabolismo , Endotelina-1/metabolismo , Síndrome de Dificultad Respiratoria del Recién Nacido/metabolismo , Transducción de Señal , Western Blotting , Líquido del Lavado Bronquioalveolar/química , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/patología , Células Cultivadas , Colon/metabolismo , Colon/patología , Complemento C5a/genética , Complemento C5a/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Endotelina-1/genética , Femenino , Fibrosis , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica , Recién Nacido , Pulmón/metabolismo , Pulmón/patología , Masculino , Microscopía Fluorescente , Miofibroblastos/metabolismo , Miofibroblastos/patología , Receptor de Anafilatoxina C5a , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Síndrome de Dificultad Respiratoria del Recién Nacido/genética , Síndrome de Dificultad Respiratoria del Recién Nacido/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trombina/genética , Trombina/metabolismo , Tromboplastina/genética , Tromboplastina/metabolismo
16.
Biomedicines ; 11(8)2023 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-37626601

RESUMEN

Childhood asthma is a chronic inflammatory airway disorder that can drive tissue remodeling. Neutrophils are amongst the most prominent inflammatory cells contributing to disease manifestations and may exert a potent role in the progression of inflammation to fibrosis. However, their role in asthma exacerbation is still understudied. Here, we investigate the association between neutrophil extracellular traps (NETs) and lung fibroblasts in childhood asthma pathophysiology using serum samples from pediatric patients during asthma exacerbation. Cell-based assays and NETs/human fetal lung fibroblast co-cultures were deployed. Increased levels of NETs and interleukin (IL)-17A were detected in the sera of children during asthma exacerbation. The in vitro stimulation of control neutrophils using the sera from pediatric patients during asthma exacerbation resulted in IL-17A-enriched NET formation. The subsequent co-incubation of lung fibroblasts with in vitro-generated IL-17A-enriched NETs led fibroblasts to acquire a pre-fibrotic phenotype, as assessed via enhanced CCN2 expression, migratory/healing capacity, and collagen release. These data uncover the important pathogenic role of the NET/IL-17A axis in asthma exacerbation, linking lung inflammation to fibroblast dysfunction and fibrosis.

18.
Arthritis Rheum ; 63(11): 3586-97, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21834070

RESUMEN

OBJECTIVE: The extrinsic coagulation cascade is involved in the fibrotic process, via thrombin-dependent induction of CCN2 (connective tissue growth factor) expression. Given the previously reported activation of the coagulation system in systemic sclerosis (SSc), we undertook the present study to investigate the involvement of cross-talk between the tissue factor (TF)-thrombin axis and endothelin 1 (ET-1) signaling in the fibrotic activity of SSc. METHODS: Human colonic myofibroblasts (HCMFs) from 6 patients with SSc and gastrointestinal symptoms and from 6 control subjects were isolated and cultured under various conditions. Messenger RNA and protein levels of TF, CCN2, and endothelin receptor A (ET(A) ) were investigated. Collagen production and migratory activity of HCMFs were further assessed. RESULTS: HCMFs from SSc patients demonstrated increased basal CCN2 production, collagen deposition, and migration rate, in a thrombin-dependent manner. Increased TF expression was also observed in SSc HCMFs. Subsequent activation of the extrinsic coagulation system resulted in thrombin-dependent enhancement of ET(A) expression. ET(A) overexpression led to further increases in both TF expression and fibrotic activity in HCMFs. Moreover, inhibition of ET-1 signaling by bosentan abolished the TF-mediated fibrotic capacity of HCMFs. CONCLUSION: Tissue factor-thrombin signaling is involved in the increased fibrotic activity of HCMFs from patients with SSc. Moreover, the up-regulation of ET(A) expression by thrombin and the effect of ET-1 in the induction of TF expression indicate an amplification loop for enhanced collagen deposition. Therapeutic interventions targeting the extrinsic coagulation system or ET-1 signaling may provide clinical benefit by breaking this vicious circle.


Asunto(s)
Colon/metabolismo , Miofibroblastos/metabolismo , Receptor de Endotelina A/metabolismo , Esclerodermia Sistémica/metabolismo , Trombina/metabolismo , Tromboplastina/metabolismo , Movimiento Celular , Proliferación Celular , Colágeno Tipo I/metabolismo , Colon/patología , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Fibrosis , Humanos , Miofibroblastos/patología , Receptor PAR-1/metabolismo , Esclerodermia Sistémica/patología , Transducción de Señal , Regulación hacia Arriba
19.
Mol Biol Rep ; 39(5): 5503-10, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22187344

RESUMEN

Familial Mediterranean fever (FMF) is an autoinflammatory disease, characterized by MEFV gene mutations and self-limited recurrent episodes of fever and localized serositis. Complement system is a key regulator of the inflammatory process. The aim of this study was to investigate the genetic alterations and mRNA expression pattern of C5aR and C5L2 genes in neutrophils from attack-free FMF patients. No mutations were observed in the two receptors' genes, while the genetic alteration observed in the C5aR1 gene was identified as N279 K polymorphic variant. Furthermore, lower mRNA expression of C5L2 gene was observed in neutrophils from FMF patients compared to control subjects. The binding capacity of rhC5a and the ability to produce reactive oxygen species was similar in neutrophils from healthy subjects and FMF patients and independent of the presence of N279 K polymorphism or mRNA expression of C5L2.


Asunto(s)
Fiebre Mediterránea Familiar/genética , Fiebre Mediterránea Familiar/patología , Predisposición Genética a la Enfermedad , Neutrófilos/metabolismo , Neutrófilos/patología , Receptores de Quimiocina/genética , Receptores de Complemento/genética , Adolescente , Adulto , Secuencia de Bases , Niño , Preescolar , Proteínas del Citoesqueleto/genética , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación/genética , Polimorfismo de Longitud del Fragmento de Restricción/genética , Polimorfismo de Nucleótido Simple/genética , Pirina , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor de Anafilatoxina C5a
20.
Arthritis Res Ther ; 24(1): 195, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35974380

RESUMEN

BACKGROUND: Accumulating evidence supports the role of monocytes and neutrophils in radiographic axSpA (r-axSpA). Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a growth factor for both leukocyte lineages and a pro-inflammatory cytokine activating myeloid cells and promoting osteoclastogenesis. It acts through the JAK-STAT pathway. We measured serum GM-CSF and markers of bone metabolism in patients with r-axSpA before and after anti-TNF treatment. METHODS: Patients with active r-axSpA despite treatment with NSAIDs, all eligible for treatment with a biologic agent, were recruited. Healthy donors were sampled as controls. Serum was collected before (baseline) and after 4-6 months (follow-up) of anti-TNF treatment and the following molecules were measured with ELISA: GM-CSF, sclerostin (SOST), and dickkopf-1 (Dkk-1). RESULTS: Twelve r-axSpA patients (7 males, 5 females, median age 37 years) with a median disease duration of 1 year and 16 age- and sex-matched controls were included. At baseline, patients had mean BASDAI 6.3±2 and ASDAS 3.2±0.7, which decreased to 4.1±1.7 and 2.2±0.6 at follow-up, respectively. At baseline, r-axSpA patients had significantly higher mean serum levels of GM-CSF (150 vs 62pg/ml, p=0.049), significantly lower Dkk-1 (1228 vs 3052pg/ml, p=0.001), but similar levels of SOST (369 vs 544pg/ml, p=0.144) compared to controls. Anti-TNF treatment did not affect GM-CSF, Dkk-1, or SOST levels. Spearman correlation analysis showed that GM-CSF correlated positively with ASDAS at baseline (r=0.61, p=0.039), while no correlations were identified between bone markers (Dkk-1, SOST) on one hand and GM-CSF or disease activity indices on the other. CONCLUSIONS: GM-CSF is increased in patients with active AS and strongly correlates with disease activity. TNF inhibition does not affect GM-SCF levels, despite improving disease activity. GM-CSF may represent an important pathway responsible for residual inflammation during TNF blockade, but also a potential target of JAK inhibitors, explaining their efficacy in r-axSpA.


Asunto(s)
Espondiloartritis Axial , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Inhibidores del Factor de Necrosis Tumoral , Adulto , Espondiloartritis Axial/sangre , Espondiloartritis Axial/tratamiento farmacológico , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/sangre , Humanos , Quinasas Janus , Masculino , Factores de Transcripción STAT , Transducción de Señal , Inhibidores del Factor de Necrosis Tumoral/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA